1
|
Oryan A, Afzali SA, Maffulli N. Manipulation of signaling pathways in bone tissue engineering and regenerative medicine: Current knowledge, novel strategies, and future directions. Injury 2024; 55:111976. [PMID: 39454294 DOI: 10.1016/j.injury.2024.111976] [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: 04/06/2024] [Revised: 09/21/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
During osteogenesis, a large number of bioactive molecules, macromolecules, cells, and cellular signals are activated to induce bone growth and development. The activation of molecular pathways leads to the occurrence of cellular events, ultimately resulting in observable changes. Therefore, in the studies of bone tissue engineering and regenerative medicine, it is essential to target fundamental events to exploit the mechanisms involved in osteogenesis. In this context, signaling pathways are activated during osteogenesis and trigger the activation of numerous other processes involved in osteogenesis. Direct influence of signaling pathways should allow to manipulate the signaling pathways themselves and impact osteogenesis. A combination of sequential cascades takes place to drive the progression of osteogenesis. Also, the occurrence of these processes and, more generally, cellular and molecular processes related to osteogenesis necessitate the presence of transcription factors and their activity. The present review focuses on outlining several signaling pathways and transcription factors influencing the development of osteogenesis, and describes various methods of their manipulation to induce and enhance bone formation.
Collapse
Affiliation(s)
- Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Seyed Ali Afzali
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Nicola Maffulli
- Department of Orthopaedic and Trauma Surgery, Faculty of Medicine and Psychology, Sant'Andrea Hospital Sapienza University of Rome, Rome, Italy; Centre for Sport and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK; Faculty of Medicine, School of Pharmacy and Bioengineering, Keele University, Stoke on Trent ST47QB, UK
| |
Collapse
|
2
|
Guo Q, Zhai Q, Ji P. The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy-Potential Implications in the Treatment of Osteogenesis Imperfecta. Pharmaceuticals (Basel) 2024; 17:1297. [PMID: 39458939 PMCID: PMC11510265 DOI: 10.3390/ph17101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Osteogenesis imperfecta (OI) is a hereditary disorder characterized by bones that are fragile and prone to breaking. The efficacy of existing therapies for OI is limited, and they are associated with potentially harmful side effects. OI is primarily due to a mutation of collagen type I and hence impairs bone regeneration. Mesenchymal stem cell (MSC) therapy is an attractive strategy to take advantage of the potential benefits of these multipotent stem cells to address the underlying molecular defects of OI by differentiating osteoblasts, paracrine effects, or immunomodulation. The maintenance of mitochondrial homeostasis is an essential component for improving the curative efficacy of MSCs in OI by affecting the differentiation, signaling, and immunomodulatory functions of MSCs. In this review, we highlight the MSC-based therapy pathway in OI and introduce the MSC regulation mechanism by mitochondrial homeostasis. Strategies aiming to modulate the metabolism and reduce the oxidative stress, as well as innovative strategies based on the use of compounds (resveratrol, NAD+, α-KG), antioxidants, and nanomaterials, are analyzed. These findings may enable the development of new strategies for the treatment of OI, ultimately resulting in improved patient outcomes.
Collapse
Affiliation(s)
- Qingling Guo
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| | - Qiming Zhai
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| | - Ping Ji
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| |
Collapse
|
3
|
Sun L, Chen J, Li LJ, Li L. Similarity-based metric analysis approach for predicting osteogenic differentiation correlation coefficients and discovering the novel osteogenic-related gene FOXA1 in BMSCs. PeerJ 2024; 12:e18068. [PMID: 39308804 PMCID: PMC11416762 DOI: 10.7717/peerj.18068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Background As a powerful tool, bioinformatics analysis is playing an increasingly important role in many fields. Osteogenic differentiation is a complex biological process involving the fine regulation of numerous genes and signaling pathways. Method Osteogenic differentiation-related genes are collected from the online databases. Then, we proposed two indexes Jaccard similarity and Sorensen-Dice similarity to measure the topological relevance of genes in the human PPI network. Furthermore, we selected three pathways involving osteoblast-related transcription factors, osteoblast differentiation, and RUNX2 regulation of osteoblast differentiation for investigation. Subsequently, we performed functional a enrichment analysis of these top-ranked genes to check whether these candidate genes identified by similarity-based metrics are enriched in some specific biological functions and states. we performed a permutation test to investigate the similarity score with four well-known osteogenic differentiation-related pathways including hedgehog signaling pathway, BMP signaling, ERK pathway, and Wnt signaling pathway to check whether these osteogenic differentiation-related pathways can be regulated by FOXA1. Lentiviral transfection was used to knockdown and overexpress gene FOXA1 in human bone mesenchymal stem cells (hBMSCs). Alkaline phosphatase (ALP) staining and Alizarin Red staining (ARS) were employed to investigate osteogenic differentiation of hBMSCs. Result After data collection, human PPI network involving 19,344 genes is included in our analysis. After simplifying, we used Jaccard and Sorensen-Dice similarity to identify osteogenic differentiation-related genes and integrated into a final similarity matrix. Furthermore, we calculated the sum of similarity scores with these osteogenic differentiation-related genes for each gene and found 337 osteogenic differentiation-related genes are involved in our analysis. We selected three pathways involving osteoblast-related transcription factors, osteoblast differentiation, and RUNX2 regulation of osteoblast differentiation for investigation and performed functional enrichment analysis of these top-ranked 50 genes. The results collectively demonstrate that these candidate genes can indeed capture osteogenic differentiation-related features of hBSMCs. According to the novel analyzing method, we found that these four pathways have significantly higher similarity with FOXA1 than random noise. Moreover, knockdown FOXA1 significantly increased the ALP activity and mineral deposits. Furthermore, overexpression of FOXA1 dramatically decreased the ALP activity and mineral deposits. Conclusion In summary, this study showed that FOXA1 is a novel significant osteogenic differentiation-related transcription factor. Moreover, our study has tightly integrated bioinformatics analysis with biological knowledge, and developed a novel method for analyzing the osteogenic differentiation regulatory network.
Collapse
Affiliation(s)
- Lingtong Sun
- Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Juan Chen
- Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Li Jun Li
- Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lingdi Li
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Yi L, Han N, Li Z, Jiang H, Cao Z. Relaxin-2 promotes osteoblastic differentiation mediated by epidermal growth factor and epidermal growth factor receptor signaling. Biotechnol Appl Biochem 2024. [PMID: 39219221 DOI: 10.1002/bab.2661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Loss of osteogenic differentiation potential of osteoblasts has been associated with the pathogenesis of osteoporosis. Thus, stimulation of osteoblastic differentiation is a therapeutic strategy for osteoporosis. Relaxin-2 is a peptide hormone with potent biological functions. However, the effects of Relaxin-2 in osteoblastic differentiation and osteoporosis have not been reported before. Here, we report a novel physiological role of Relaxin-2 in promoting osteoblastic differentiation and mineralization of MC3T3-E1 cells. Our results indicate that exposure to Relaxin-2 upregulated the expression, and elevated the activity of alkaline phosphatase (ALP) when MC3T3-E1 cells were cultured in osteogenic differentiation medium (OM). Additionally, Relaxin-2 upregulated the mRNA levels of osteocalcin (ocn), osteopontin (opn), and collagen type I alpha 1 (Col1a1). The alizarin red S staining assay revealed that Relaxin-2 promoted the mineralization of MC3T3-E1 cells. We also found that Relaxin-2 increased the expression of Runx-2 as well as the epidermal growth factor (EGF) and epidermal growth factor receptor (EGFR). Importantly, silencing of EGF abolished the effects of Relaxin-2 in osteoblastic differentiation and related gene expression. These findings suggest that Relaxin-2 stimulates osteogenic differentiation through activating EGF/EGFR signaling.
Collapse
Affiliation(s)
- Lankai Yi
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Ning Han
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhong Li
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Housen Jiang
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| | - Zhenhao Cao
- Department of Hand, Foot, and Orthopedics Surgery, Weifang People's Hospital, Weifang, Shandong Province, China
| |
Collapse
|
5
|
Prins CM, Ceylan M, Hogervorst JMA, Jansen IDC, Schimmel IM, Schoenmaker T, de Vries TJ. Osteogenic differentiation of periodontal ligament fibroblasts inhibits osteoclast formation. Eur J Cell Biol 2024; 103:151440. [PMID: 38954934 DOI: 10.1016/j.ejcb.2024.151440] [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: 03/26/2024] [Revised: 06/02/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
One of the deficits of knowledge on bone remodelling, is to what extent cells that are driven towards osteogenic differentiation can contribute to osteoclast formation. The periodontal ligament fibroblast (PdLFs) is an ideal model to study this, since they play a role in osteogenesis, and can also orchestrate osteoclastogenesis.when co-cultured with a source of osteoclast-precursor such as peripheral blood mononuclear cells (PBMCs). Here, the osteogenic differentiation of PdLFs and the effects of this process on the formation of osteoclasts were investigated. PdLFs were obtained from extracted teeth and exposed to osteogenic medium for 0, 7, 14, or 21 out of 21 days. After this 21-day culturing period, the cells were co-cultured with peripheral blood mononuclear cells (PBMCs) for an additional 21 days to study osteoclast formation. Alkaline phosphatase (ALP) activity, calcium concentration, and gene expression of osteogenic markers were assessed at day 21 to evaluate the different stages of osteogenic differentiation. Alizarin red staining and scanning electron microscopy were used to visualise mineralisation. Tartrate-resistant acid phosphatase (TRAcP) activity, TRAcP staining, multinuclearity, the expression of osteoclastogenesis-related genes, and TNF-α and IL-1β protein levels were assessed to evaluate osteoclastogenesis. The osteogenesis assays revealed that PdLFs became more differentiated as they were exposed to osteogenic medium for a longer period of time. Mineralisation by these osteogenic cells increased with the progression of differentiation. Culturing PdLFs in osteogenic medium before co-culturing them with PMBCs led to a significant decrease in osteoclast formation. qPCR revealed significantly lower DCSTAMP expression in cultures that had been supplemented with osteogenic medium. Protein levels of osteoclastogenesis stimulator TNF-α were also lower in these cultures. The present study shows that the osteogenic differentiation of PdLFs reduces the osteoclastogenic potential of these cells. Immature cells of the osteoblastic lineage may facilitate osteoclastogenesis, whereas mature mineralising cells may suppress the formation of osteoclasts. Therefore, mature and immature osteogenic cells may have different roles in maintaining bone homeostasis.
Collapse
Affiliation(s)
- Caya M Prins
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands; Amsterdam University College (University of Amsterdam and Vrije Universiteit), Amsterdam, the Netherlands
| | - Merve Ceylan
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Jolanda M A Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Ineke D C Jansen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Irene M Schimmel
- Department of Medical Biology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands
| | - Teun J de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Chan HTL, Chan KM, Abhreet-Kaur, Sam SW, Chan SW. A Review of the Pharmacological Effects of Solanum muricatum Fruit (Pepino Melon). Foods 2024; 13:2740. [PMID: 39272505 PMCID: PMC11394486 DOI: 10.3390/foods13172740] [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: 07/23/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Solanaceae, commonly known as nightshade, is a diverse family of flowering plants comprising around 90 genera and an estimated 3000-4000 species. Solanaceae spp. and its various fruits, including pepino (Solanum muricatum), commonly known as pepino melon, are widely recognized by the public for their nutritional value and pharmacological effects. Pepino melon, in particular, is often enjoyed as a fresh dessert or salad due to its juicy flesh. Given its beneficial properties, the potential of pepino melon to be developed as a functional food has been extensively studied. This review aims to provide a comprehensive summary of the reported pharmacological effects of the active compounds found in pepino plant and melon. Among these compounds, polyphenols, notably quercetin, and vitamin C have demonstrated notable antioxidant properties such as scavenging free radicals, effectively protecting against free-radical damage. Moreover, these active ingredients provide pepino with anti-inflammatory properties by inhibiting the expression of proinflammatory cytokines and enzymes, thereby reducing nitric oxide production. Additionally, they have shown promise in selectively targeting cancer cells, exhibiting anti-cancer properties. Furthermore, the active compounds such as quercetin in pepino have been associated with anti-diabetic effects, improving insulin sensitivity and inhibiting insulin resistance. Overall, this review highlights the diverse and significant pharmacological effects of the active compounds found in pepino melon, emphasizing its potential as a valuable functional food.
Collapse
Affiliation(s)
- Hei-Tung Lydia Chan
- Department of Food and Health Science, Technological and Higher Education Institute of Hong Kong, Hong Kong
| | - Ka-Man Chan
- Department of Food and Health Science, Technological and Higher Education Institute of Hong Kong, Hong Kong
| | - Abhreet-Kaur
- Department of Food and Health Science, Technological and Higher Education Institute of Hong Kong, Hong Kong
| | - Sze-Wing Sam
- Department of Food and Health Science, Technological and Higher Education Institute of Hong Kong, Hong Kong
| | - Shun-Wan Chan
- Department of Food and Health Science, Technological and Higher Education Institute of Hong Kong, Hong Kong
| |
Collapse
|
7
|
Arya PN, Saranya I, Selvamurugan N. RUNX2 regulation in osteoblast differentiation: A possible therapeutic function of the lncRNA and miRNA-mediated network. Differentiation 2024:100803. [PMID: 39089986 DOI: 10.1016/j.diff.2024.100803] [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: 05/09/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Osteogenic differentiation is a crucial process in the formation of the skeleton and the remodeling of bones. It relies on a complex system of signaling pathways and transcription factors, including Runt-related transcription factor 2 (RUNX2). Non-coding RNAs (ncRNAs) control the bone-specific transcription factor RUNX2 through post-transcriptional mechanisms to regulate osteogenic differentiation. The most research has focused on microRNAs (miRNAs) and long ncRNAs (lncRNAs) in studying how they regulate RUNX2 for osteogenesis in both normal and pathological situations. This article provides a concise overview of the recent advancements in understanding the critical roles of lncRNA/miRNA/axes in controlling the expression of RUNX2 during bone formation. The possible application of miRNAs and lncRNAs as therapeutic agents for the treatment of disorders involving the bones and bones itself is also covered.
Collapse
Affiliation(s)
- Pakkath Narayanan Arya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Iyyappan Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
8
|
Kang Y, Na J, Karima G, Amirthalingam S, Hwang NS, Kim HD. Mesenchymal Stem Cell Spheroids: A Promising Tool for Vascularized Tissue Regeneration. Tissue Eng Regen Med 2024; 21:673-693. [PMID: 38578424 PMCID: PMC11187036 DOI: 10.1007/s13770-024-00636-2] [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: 01/05/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are undifferentiated cells that can differentiate into specific cell lineages when exposed to the right conditions. The ability of MSCs to differentiate into particular cells is considered very important in biological research and clinical applications. MSC spheroids are clusters of MSCs cultured in three dimensions, which play an important role in enhancing the proliferation and differentiation of MSCs. MSCs can also participate in vascular formation by differentiating into endothelial cells and secreting paracrine factors. Vascularization ability is essential in impaired tissue repair and function recovery. Therefore, the vascularization ability of MSCs, which enhances angiogenesis and accelerates tissue healing has made MSCs a promising tool for tissue regeneration. However, MSC spheroids are a relatively new research field, and more research is needed to understand their full potential. METHODS In this review, we highlight the importance of MSC spheroids' vascularization ability in tissue engineering and regenerative medicine while providing the current status of studies on the MSC spheroids' vascularization and suggesting potential future research directions for MSC spheroids. RESULTS Studies both in vivo and in vitro have demonstrated MSC spheroids' capacity to develop into endothelial cells and stimulate vasculogenesis. CONCLUSION MSC spheroids show potential to enhance vascularization ability in tissue regeneration. Yet, further research is required to comprehensively understand the relationship between MSC spheroids and vascularization mechanisms.
Collapse
Affiliation(s)
- Yoonjoo Kang
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Jinwoo Na
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwan D Kim
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, 27469, Republic of Korea.
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea.
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
9
|
Valenti MT, Zerlotin R, Cominacini M, Bolognin S, Grano M, Dalle Carbonare L. Exploring the Role of Circular RNA in Bone Biology: A Comprehensive Review. Cells 2024; 13:999. [PMID: 38920630 PMCID: PMC11201515 DOI: 10.3390/cells13120999] [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: 05/09/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Circular RNAs (circRNAs) have emerged as pivotal regulators of gene expression with diverse roles in various biological processes. In recent years, research into circRNAs' involvement in bone biology has gained significant attention, unveiling their potential as novel regulators and biomarkers in bone-related disorders and diseases. CircRNAs, characterized by their closed-loop structure, exhibit stability and resistance to degradation, underscoring their functional significance. In bone tissue, circRNAs are involved in critical processes such as osteogenic differentiation, osteoclastogenesis, and bone remodeling through intricate molecular mechanisms including microRNA regulation. Dysregulated circRNAs are associated with various bone disorders, suggesting their potential as diagnostic and prognostic biomarkers. The therapeutic targeting of these circRNAs holds promise for addressing bone-related conditions, offering new perspectives for precision medicine. Thus, circRNAs constitute integral components of bone regulatory networks, impacting both physiological bone homeostasis and pathological conditions. This review provides a comprehensive overview of circRNAs in bone biology, emphasizing their regulatory mechanisms, functional implications, and therapeutic potential.
Collapse
Affiliation(s)
- Maria Teresa Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy
| | - Roberta Zerlotin
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.G.)
| | - Mattia Cominacini
- Department of Engineering for the Innovation Medicine, University of Verona, 37100 Verona, Italy; (M.C.); (L.D.C.)
| | - Silvia Bolognin
- MERLN Institute, Maastricht University, Universiteitssingel 40, 6229 ET Maastricht, The Netherlands;
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari, 70124 Bari, Italy; (R.Z.); (M.G.)
| | - Luca Dalle Carbonare
- Department of Engineering for the Innovation Medicine, University of Verona, 37100 Verona, Italy; (M.C.); (L.D.C.)
| |
Collapse
|
10
|
Ke KX, Gao X, Liu L, He WG, Jiang Y, Long CB, Zhong G, Xu ZH, Deng ZL, He BC, Hu N. Leptin attenuates the osteogenic induction potential of BMP9 by increasing β-catenin malonylation modification via Sirt5 down-regulation. Aging (Albany NY) 2024; 16:7870-7888. [PMID: 38709288 PMCID: PMC11131982 DOI: 10.18632/aging.205790] [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: 11/27/2023] [Accepted: 03/29/2024] [Indexed: 05/07/2024]
Abstract
BMP9 has demonstrated significant osteogenic potential. In this study, we investigated the effect of Leptin on BMP9-induced osteogenic differentiation. Firstly, we found Leptin was decreased during BMP9-induced osteogenic differentiation and serum Leptin concentrations were increased in the ovariectomized (OVX) rats. Both in vitro and in vivo, exogenous expression of Leptin inhibited the process of osteogenic differentiation, whereas silencing Leptin enhanced. Exogenous Leptin could increase the malonylation of β-catenin. However, BMP9 could increase the level of Sirt5 and subsequently decrease the malonylation of β-catenin; the BMP9-induced osteogenic differentiation was inhibited by silencing Sirt5. These data suggested that Leptin can inhibit the BMP9-induced osteogenic differentiation, which may be mediated through reducing the activity of Wnt/β-catenin signalling via down-regulating Sirt5 to increase the malonylation level of β-catenin partly.
Collapse
Affiliation(s)
- Kai-Xin Ke
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Xiang Gao
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Department of Orthopaedics, The second affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Lu Liu
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Wen-Ge He
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Department of Orthopaedics, The first affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Yue Jiang
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Cheng-Bin Long
- Department of Orthopaedics, The first affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Department of Orthopaedics, Bishan Hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Gan Zhong
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Department of Orthopaedics, The first affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Zheng-Hao Xu
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Department of Orthopaedics, The first affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Zhong-Liang Deng
- Department of Orthopaedics, The second affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Bai-Cheng He
- Department of Pharmacology, School of Pharmacy, Chongqing Medical University, Chongqing 400016, People’s Republic of China
- Key Laboratory of Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing 400016, People’s Republic of China
| | - Ning Hu
- Department of Orthopaedics, The first affiliated hospital of Chongqing Medical University, Chongqing 400016, People’s Republic of China
| |
Collapse
|
11
|
Çakmak A, Fuerkaiti S, Karagüzel D, Karaaslan Ç, Gümüşderelioğlu M. Enhanced Osteogenic Potential of Noggin Knockout C2C12 Cells on BMP-2 Releasing Silk Scaffolds. ACS Biomater Sci Eng 2023; 9:6175-6185. [PMID: 37796024 PMCID: PMC10646847 DOI: 10.1021/acsbiomaterials.3c00506] [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: 04/18/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023]
Abstract
The CRISPR/Cas9 mechanism offers promising therapeutic approaches for bone regeneration by stimulating or suppressing critical signaling pathways. In this study, we aimed to increase the activity of BMP-2 signaling through knockout of Noggin, thereby establishing a synergistic effect on the osteogenic activity of cells in the presence of BMP-2. Since Noggin is an antagonist expressed in skeletal tissues and binds to subunits of bone morphogenetic proteins (BMPs) to inhibit osteogenic differentiation, here Noggin expression was knocked out using the CRISPR/Cas9 system. In accordance with this purpose, C2C12 (mouse myoblast) cells were transfected with CRISPR/Cas9 plasmids. Transfection was achieved with Lipofectamine and confirmed with intense fluorescent signals in microscopic images and deletion in target sequence in Sanger sequencing analysis. Thus, Noggin knockout cells were identified as a new cell source for tissue engineering studies. Then, the transfected cells were seeded on highly porous silk scaffolds bearing BMP-2-loaded silk nanoparticles (30 ng BMP-2/mg silk nanoparticle) in the size of 288 ± 62 nm. BMP-2 is released from the scaffolds in a controlled manner for up to 60 days. The knockout of Noggin by CRISPR/Cas9 was found to synergistically promote osteogenic differentiation in the presence of BMP-2 through increased Coll1A1 and Ocn expression and mineralization. Gene editing of Noggin and BMP-2 increased almost 2-fold Col1A1 expression and almost 3-fold Ocn expression compared to the control group. Moreover, transfected cells produced extracellular matrix (ECM) containing collagen fibers on the scaffolds and mineral-like structures were formed on the fibers. In addition, mineralization characterized by intense Alizarin red staining was detected in transfected cells cultured in the presence of BMP-2, while the other groups did not exhibit any mineralized areas. As has been demonstrated in this study, the CRISPR/Cas9 mechanism has great potential for obtaining new cell sources to be used in tissue engineering studies.
Collapse
Affiliation(s)
- Anıl
Sera Çakmak
- Department
of Chemical Engineering, Hacettepe University, 06800 Ankara, Turkey
| | - Sümeyra Fuerkaiti
- Division
of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
| | - Dilara Karagüzel
- Department
of Biology, Molecular Biology Section, Hacettepe
University, 06800 Ankara, Turkey
| | - Çağatay Karaaslan
- Division
of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
- Department
of Biology, Molecular Biology Section, Hacettepe
University, 06800 Ankara, Turkey
| | - Menemşe Gümüşderelioğlu
- Department
of Chemical Engineering, Hacettepe University, 06800 Ankara, Turkey
- Division
of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, 06800 Ankara, Turkey
| |
Collapse
|
12
|
Ebrahimi F, Pirouzmand F, Cosme Pecho RD, Alwan M, Yassen Mohamed M, Ali MS, Hormozi A, Hasanzadeh S, Daei N, Hajimortezayi Z, Zamani M. Application of mesenchymal stem cells in regenerative medicine: A new approach in modern medical science. Biotechnol Prog 2023; 39:e3374. [PMID: 37454344 DOI: 10.1002/btpr.3374] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
Mesenchymal Stem Cells (MSCs) are non-hematopoietic and multipotent stem cells, which have been considered in regenerative medicine. These cells are easily separated from different sources, such as bone marrow (BM), umbilical cord (UC), adipose tissue (AT), and etc. MSCs have the differentiation capability into chondrocytes, osteocytes, and adipocytes; This differentiation potential along with the paracrine properties have made them a key choice for tissue repair. MSCs also have various advantages over other stem cells, which is why they have been extensively studied in recent years. The effectiveness of MSCs-based therapies depend on several factors, including differentiation status at the time of use, concentration per injection, delivery method, the used vehicle, and the nature and extent of the damage. Although, MSCs have emerged promising sources for regenerative medicine, there are potential risks regarding their safety in their clinical use, including tumorigenesis, lack of availability, aging, and sensitivity to toxic environments. In this study, we aimed to discuss how MSCs may be useful in treating defects and diseases. To this aim, we will review recent advances of MSCs action mechanisms in regenerative medicine, as well as the most recent clinical trials. We will also have a brief overview of MSCs resources, differences between their sources, culture conditions, extraction methods, and clinical application of MSCs in various fields of regenerative medicine.
Collapse
Affiliation(s)
- Faezeh Ebrahimi
- Medical Laboratory, Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Farzaneh Pirouzmand
- Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | | | - Mariam Alwan
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | | | - Arezoo Hormozi
- Medical Laboratory, Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Sajedeh Hasanzadeh
- Medical Laboratory, Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Narges Daei
- Medical Laboratory, Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zahra Hajimortezayi
- Medical Laboratory, Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Majid Zamani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
13
|
Mo L, Chen L, Wan Y, Huang H, Mo L, Zhu W, Yang G, Li Z, Wei Q, Song J, Yang X. An aqueous extract of Prunella vulgaris L. ameliorates cadmium-induced bone loss by promoting osteogenic differentiation in female rats. Food Chem Toxicol 2023; 180:114005. [PMID: 37640280 DOI: 10.1016/j.fct.2023.114005] [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: 07/12/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
Cadmium (Cd) causes bone loss, concerning inhibiting osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Prunella vulgaris L. (PV) has the potential for promoting osteogenic differentiation, but its influence on Cd-induced bone loss is unclear. This study investigated the effect of PV aqueous extract (PVE) on Cd-induced bone loss and its underlying mechanisms. Eight-week-old female SD rats were randomly assigned into four groups and treated for 16 weeks: Control, Cd (50 mg/L of Cd chloride), Cd + PV Low (125 mg/kg bw of PVE), and Cd + PV High (250 mg/kg bw of PVE). PV ameliorated femoral bone loss in Cd-treated rats manifested as increases in bone mineral density, bone volume, trabecular thickness, number, and area, and decreases in trabecular separation. Compared with Cd group, PV-treatment groups had higher serum levels of bone formation markers (ALP, BGP). Additionally, in PV-treatment groups, expressions of bone formation markers (Osterix, Runx2) and molecules involved in osteogenic differentiation signal pathway BMP/Smad (BMP4, Smad1/5/9) in the tibia of rats and isolated rat primary BMSCs were upregulated. These results suggest that PV alleviates Cd-induced bone loss by promoting osteogenic differentiation, which is likely associated with BMP/Smad pathway.
Collapse
Affiliation(s)
- Lijun Mo
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Linquan Chen
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Yu Wan
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Haibin Huang
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Lifen Mo
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Wei Zhu
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, PR China
| | - Guangyu Yang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, PR China
| | - Ziyin Li
- Affiliated Dongguan Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523059, PR China
| | - Qinzhi Wei
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China
| | - Jia Song
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China.
| | - Xingfen Yang
- Food Safety and Health Research Center, NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
14
|
Abnosi MH, Sargolzaei J, Nazari F. Gallic Acid Ameliorates Cadmium Effect on Osteogenesis by Activation of Alkaline Phosphatase and Collagen Synthesis. CELL JOURNAL 2023; 25:603-612. [PMID: 37718763 PMCID: PMC10520984 DOI: 10.22074/cellj.2023.1999110.1263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/17/2023] [Accepted: 07/26/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVE We previously reported that cadmium (Cd) inhibits osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). In addition, gallic acid (GA) improves BMSC differentiation. Here, we aim to study the ability of GA to prevent osteogenic inhibition induced by Cd. MATERIALS AND METHODS In this experimental study, BMSCs were extracted and purified from Wistar rats and their viability was determined in the presence of Cd and GA. The results indicated that 1.5 μM Cd and 0.25 μM of GA were appropriate for further investigation. After 20 days in osteogenic medium, matrix production was analysed by alizarin red, calcium content, and alkaline phosphatase (ALP) activity. Osteogenic-related genes and collagen 1A1 (COL1A1) protein expressions were investigated. The preventive effect of GA on oxidative stress and metabolic change induced by Cd was estimated. RESULTS GA counteracted the inhibitory effect of Cd on matrix production and significantly (P=0.0001) improved the osteogenic differentiation ability of BMSCs. Also, GA prevented the toxic effect of Cd on osteogenic-related gene expressions and nullified the reducing effect of Cd on COL1Al and ALP activity. A significant reduction (P=0.0001) in malondialdehyde and lactic acid concentration showed that GA counteracted both oxidative stress and metabolic changes caused by Cd. CONCLUSION GA prevented the toxic effect of Cd, an environmental pollutant and a factor in osteoporosis.
Collapse
Affiliation(s)
| | - Javad Sargolzaei
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Farshid Nazari
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| |
Collapse
|
15
|
Xia SL, Ma ZY, Wang B, Gao F, Guo SY, Chen XH. Icariin promotes the proliferation and osteogenic differentiation of bone-derived mesenchymal stem cells in patients with osteoporosis and T2DM by upregulating GLI-1. J Orthop Surg Res 2023; 18:500. [PMID: 37454090 DOI: 10.1186/s13018-023-03998-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND The function of mesenchymal stem cells (MSCs) from patients with osteoporosis (OP) is impaired and worsens in patients with type 2 diabetes mellitus (T2DM). Icariin (ICA) is the major active flavonoid glucoside isolated from traditional Chinese herbal Epimedium pubescens, and confirmed able to improve bone mass of OP patients. OBJECTIVE To investigate the effect of ICA on the proliferation and osteogenic differentiation of bone-derived MSCs (BMSCs) from patients with OP and T2DM and uncover the potential mechanism. METHODS BMSCs were treated with ICA, and proliferation and osteogenic potency were evaluated using the 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay and detection of osteogenic markers (ALP, RUNX2, SPP1, COL1A1, and mineralized nodules) was performed. RNA sequencing and bioinformatic analysis were performed to identify differentially expressed genes (DEGs) after ICA treatment and screen proliferation- and osteogenic differentiation-related processes. Gene gain and loss were performed to confirm the role of the key candidate gene. RESULTS ICA significantly promoted the proliferation and osteogenic differentiation of BMSCs. A total of 173 DEGs were identified after ICA treatment. Six DEGs (GLI-1, IGF2, BMP6, WNT5A, PTHLH, and MAPK14) enriched in both proliferation- and osteogenic differentiation-related processes were screened; GLI-1 had the highest validated |log2FC| value. Overexpression of GLI-1 enhanced the proliferation and osteogenic differentiation of BMSCs, and knockdown of GLI-1 weakened the positive effect of ICA on BMSCs. CONCLUSION ICA promoted the proliferation and osteogenic differentiation of impaired BMSCs by upregulating GLI-1.
Collapse
Affiliation(s)
- Sheng-Li Xia
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Zi-Yuan Ma
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Bin Wang
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Feng Gao
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Sheng-Yang Guo
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China
| | - Xu-Han Chen
- Zhoupu Community Health Service Center, 163 Shenmei East Road, Pudong New Area, Shanghai, 201318, China.
| |
Collapse
|
16
|
Yu R, Han H, Chu S, Ding Y, Jin S, Wang Y, Jiang W, Liu Y, Zou Y, Wang M, Liu Q, Sun G, Jiang B, Gong Y. CUL4B orchestrates mesenchymal stem cell commitment by epigenetically repressing KLF4 and C/EBPδ. Bone Res 2023; 11:29. [PMID: 37268647 DOI: 10.1038/s41413-023-00263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 06/04/2023] Open
Abstract
Dysregulated lineage commitment of mesenchymal stem cells (MSCs) contributes to impaired bone formation and an imbalance between adipogenesis and osteogenesis during skeletal aging and osteoporosis. The intrinsic cellular mechanism that regulates MSC commitment remains unclear. Here, we identified Cullin 4B (CUL4B) as a critical regulator of MSC commitment. CUL4B is expressed in bone marrow MSCs (BMSCs) and downregulated with aging in mice and humans. Conditional knockout of Cul4b in MSCs resulted in impaired postnatal skeletal development with low bone mass and reduced bone formation. Moreover, depletion of CUL4B in MSCs aggravated bone loss and marrow adipose accumulation during natural aging or after ovariectomy. In addition, CUL4B deficiency in MSCs reduced bone strength. Mechanistically, CUL4B promoted osteogenesis and inhibited adipogenesis of MSCs by repressing KLF4 and C/EBPδ expression, respectively. The CUL4B complex directly bound to Klf4 and Cebpd and epigenetically repressed their transcription. Collectively, this study reveals CUL4B-mediated epigenetic regulation of the osteogenic or adipogenic commitment of MSCs, which has therapeutic implications in osteoporosis.
Collapse
Affiliation(s)
- Ruiqi Yu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Hong Han
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuxian Chu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yijun Ding
- The Key Laboratory of Liquid‒Solid Structural Evolution and Processing of Materials of Ministry of Education and Institute of Liquid Metal and Casting Technology, School of Materials Science and Engineering, Shandong University, Jinan, 250012, China
| | - Shiqi Jin
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yufeng Wang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Wei Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yuting Liu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Molin Wang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Qiao Liu
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Genetics, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
17
|
Moura SR, Fernandes MJ, Santos SG, Almeida MI. Circular RNAs: Promising Targets in Osteoporosis. Curr Osteoporos Rep 2023; 21:289-302. [PMID: 37119447 PMCID: PMC10169890 DOI: 10.1007/s11914-023-00786-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE OF REVIEW Circular RNAs (circRNAs) are RNA transcripts derived from fragments of pre-messenger RNAs through a back-splicing process. An advantage that rises from their circular covalently closed conformation is their high stability, when compared with their linear counterparts. The current review focuses on the emerging roles of circRNAs in osteoporosis, including in osteogenic differentiation and osteoclastogenesis. Their potential as osteoporosis biomarkers will also be discussed. RECENT FINDINGS Although firstly described as non-coding, some of these single-stranded RNAs were recently reported to possess protein-coding capacity. On the other hand, the circRNAs exhibit cell and tissue-specific patterns at the transcriptome level in eukaryotes and are regulated throughout the development or disease progression. Even though thousands of these circular transcripts are listed and annotated, only a limited number of studies describe their biological role in bone processes. Recent evidence indicates inhibitory activator roles in both osteoblasts and osteoclasts differentiation and function. Latest screenings in the blood, plasma, or serum of osteoporosis patients support the potential for circRNA signature to be used as biomarkers in osteoporosis, but further validation is required. While intense research into circRNAs has been detailing their biological roles, there remains a need for standardization and further research to fulfil the future potential of this emerging and highly promising class of regulatory molecules.
Collapse
Affiliation(s)
- Sara Reis Moura
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria João Fernandes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Susana G Santos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| | - Maria Inês Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
18
|
Melo US, Jatzlau J, Prada-Medina CA, Flex E, Hartmann S, Ali S, Schöpflin R, Bernardini L, Ciolfi A, Moeinzadeh MH, Klever MK, Altay A, Vallecillo-García P, Carpentieri G, Delledonne M, Ort MJ, Schwestka M, Ferrero GB, Tartaglia M, Brusco A, Gossen M, Strunk D, Geißler S, Mundlos S, Stricker S, Knaus P, Giorgio E, Spielmann M. Enhancer hijacking at the ARHGAP36 locus is associated with connective tissue to bone transformation. Nat Commun 2023; 14:2034. [PMID: 37041138 PMCID: PMC10090176 DOI: 10.1038/s41467-023-37585-8] [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: 06/08/2022] [Accepted: 03/21/2023] [Indexed: 04/13/2023] Open
Abstract
Heterotopic ossification is a disorder caused by abnormal mineralization of soft tissues in which signaling pathways such as BMP, TGFβ and WNT are known key players in driving ectopic bone formation. Identifying novel genes and pathways related to the mineralization process are important steps for future gene therapy in bone disorders. In this study, we detect an inter-chromosomal insertional duplication in a female proband disrupting a topologically associating domain and causing an ultra-rare progressive form of heterotopic ossification. This structural variant lead to enhancer hijacking and misexpression of ARHGAP36 in fibroblasts, validated here by orthogonal in vitro studies. In addition, ARHGAP36 overexpression inhibits TGFβ, and activates hedgehog signaling and genes/proteins related to extracellular matrix production. Our work on the genetic cause of this heterotopic ossification case has revealed that ARHGAP36 plays a role in bone formation and metabolism, outlining first details of this gene contributing to bone-formation and -disease.
Collapse
Affiliation(s)
- Uirá Souto Melo
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany.
- Institute for Medical Genetics and Human Genetics, Charité University Medicine Berlin, 13353, Berlin, Germany.
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, 14195, Berlin, Germany
| | - Cesar A Prada-Medina
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany
| | - Elisabetta Flex
- Istituto Superiore di Sanità, Department of Oncology and Molecular Medicine, 00161, Rome, Italy
| | - Sunhild Hartmann
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany
| | - Salaheddine Ali
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany
| | - Robert Schöpflin
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany
| | - Laura Bernardini
- Cytogenetics Unit, Casa Sollievo della Sofferenza Foundation, IRCCS, 71013, San Giovanni Rotondo, Foggia, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - M-Hossein Moeinzadeh
- Max Planck Institute for Molecular Genetics, Department of Computational Molecular Biology, 14195, Berlin, Germany
| | - Marius-Konstantin Klever
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité University Medicine Berlin, 13353, Berlin, Germany
| | - Aybuge Altay
- Max Planck Institute for Molecular Genetics, Department of Computational Molecular Biology, 14195, Berlin, Germany
| | | | - Giovanna Carpentieri
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | | | - Melanie-Jasmin Ort
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, 14195, Berlin, Germany
- Julius Wolff Institute (JWI), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Marko Schwestka
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| | | | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - Alfredo Brusco
- Department of Medical Sciences, University of Torino, 10126, Torino, Italy
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, 10126, Italy
| | - Manfred Gossen
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 14513, Teltow, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| | - Dirk Strunk
- Cell Therapy Institute, Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University (PMU), 5020, Salzburg, Austria
| | - Sven Geißler
- Julius Wolff Institute (JWI), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 13353, Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353, Berlin, Germany
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany
- Institute for Medical Genetics and Human Genetics, Charité University Medicine Berlin, 13353, Berlin, Germany
| | - Sigmar Stricker
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, 14195, Berlin, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, 14195, Berlin, Germany
| | - Elisa Giorgio
- Department of Molecular Medicine, University of Pavia, 27100, Pavia, Italy.
- Medical Genetics Unit, IRCCS Mondino Foundation, 27100, Pavia, Italy.
| | - Malte Spielmann
- Max Planck Institute for Molecular Genetics, Development and Disease Group, 14195, Berlin, Germany.
- Institute of Human Genetics, University Hospitals Schleswig-Holstein, University of Lübeck and University of Kiel, Lübeck, 23562, Germany.
- DZHK (German Centre for Cardiovascular Research) Germany, partner site Hamburg, Lübeck, Kiel, Lübeck, 23562, Germany.
| |
Collapse
|
19
|
Wang H, Xu W, Chen X, Mei X, Guo Z, Zhang J. LncRNA LINC00205 stimulates osteoporosis and contributes to spinal fracture through the regulation of the miR-26b-5p/KMT2C axis. BMC Musculoskelet Disord 2023; 24:262. [PMID: 37016415 PMCID: PMC10071705 DOI: 10.1186/s12891-023-06136-z] [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: 06/29/2022] [Accepted: 01/05/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND Osteoporosis (OP) is a common bone disease marked by decreased bone strength. Increasing evidence suggests that long non-coding RNA (lncRNAs) play important roles in the occurrence and progression of OP. This study aimed to investigate the role and mechanism of LINC00205 in the osteogenic differentiation of human mesenchymal stem cells (hMSCs) and OP. METHODS Bone tissue samples were obtained from healthy controls and patients with osteoporosis with a spinal fracture (OP-Frx) or without a spinal fracture (OP-no-Frx). HMSCs were cultured and induced to undergo osteogenic differentiation. The expression of LINC00205, lysine (K)-specific methyltransferase 2C (KMT2C), and miR-26b-5p in bone tissues and cells was evaluated using western blotting and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). The effects of LINC00205, miR-26b-5p, and KMT2C on calcium deposition, alkaline phosphatase (ALP) activity, and mRNA levels of the osteogenic differentiation marker genes [ALP, osteocalcin (OCN), and runt-related transcription factor 2 (RUNX2)] were investigated using alizarin red S staining, an ALP activity assay, and qRT-PCR, respectively. Dual-luciferase reporter assay was performed to ascertain the binding relationship between miR-26b-5p and LINC00205/KMT2C. RESULTS LINC00205 and KMT2C were upregulated in patients with OP-Frx and OP-no-Frx, whereas miR-26b-5p was downregulated. Furthermore, LINC00205 and KMT2C expression decreased, whereas that of miR-26b-5p increased over time from day 7 to 21 of the osteogenic differentiation of hMSCs. The knockdown of LINC00205 and KMT2C significantly increased ALP activity, calcium deposition, and the expression of RUNX2, ALP, and OCN. In contrast, the inhibition of miR-26b-5p yielded the opposite result. These data suggest that LINC00205 inhibits the osteogenic differentiation of hMSCs by modulating the miR-26b-5p/KMT2C signaling axis. CONCLUSION LINC00205 promotes OP and is involved in spinal fractures. LINC00205 is also a potential negative regulator of the osteogenic differentiation of hMSCs.
Collapse
Affiliation(s)
- Hongtao Wang
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Weilin Xu
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Xiaoqing Chen
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Xiongfeng Mei
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China
| | - Zhonghua Guo
- Department of Orthopaedics, People's Hospital of Dongxihu District, Wuhan, 430040, Hubei, China
| | - Juan Zhang
- Department of Rehabilitation Medicine, People's Hospital of Dongxihu District, No. 48 Jinbei 1St Road, Jinghe Street, Dongxihu District, Wuhan, 430040, Hubei, China.
| |
Collapse
|
20
|
Zheng X, Wang W, Chen S, Zuo B, Li J. Transplanted mesenchymal stromal cells are unable to migrate to the bone surface and subsequently improve osteogenesis in glucocorticoid-induced osteoporosis. Cytotherapy 2023; 25:472-482. [PMID: 36863932 DOI: 10.1016/j.jcyt.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/14/2022] [Accepted: 01/07/2023] [Indexed: 03/03/2023]
Abstract
Long-term or high-dose use of glucocorticoids causes bone loss and low bone formation. We previously demonstrated that dexamethasone (Dex) administration caused the shifted differentiation balance of mesenchymal stromal cells (MSCs) to favor adipogenic lineage over osteoblastic lineage, which is one of the key mechanisms for Dex-induced osteoporosis (DIO). These findings indicate that supplementing functional allogeneic MSCs could be a therapeutic strategy for DIO. Here, we found that transplanting MSCs by intramedullary injection had little effect in promoting new bone formation. Fluorescent-labeled lineage tracing revealed that 1 week after transplantation, green fluorescent protein (GFP)-MSCs were found to migrate to the bone surface (BS) in control mice but not in DIO mice. As expected, GFP-MSCs on the BS were mostly Runx2-positive; however, GFP-MSCs located away from the BS failed to differentiate into osteoblasts. We further discovered that the levels of transforming growth factor beta 1 (TGF-β1), one of the main chemokines for MSC migration, is significantly decreased in the bone marrow fluid of DIO mice, which is insufficient to direct MSC migration. Mechanistically, Dex inhibits TGF-β1 expression by down-regulating its promoter activity, which decreases bone matrix-deposited TGF-β1 as well as active TGF-β1 released during osteoclast-mediated bone resorption. This study indicates that blocking MSC migration in osteoporotic BM contributes to bone loss and suggests that MSC mobilization to the BS may be a promising target for treating osteoporosis.
Collapse
Affiliation(s)
- Xueling Zheng
- Department of Cell Biology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Wanyuji Wang
- Department of Cell Biology, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Sisi Chen
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Zuo
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiao Li
- Department of Cell Biology, Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
21
|
Zeng C, Wang S, Chen F, Wang Z, Li J, Xie Z, Ma M, Wang P, Shen H, Wu Y. Alpinetin alleviates osteoporosis by promoting osteogenic differentiation in BMSCs by triggering autophagy via PKA/mTOR/ULK1 signaling. Phytother Res 2023; 37:252-270. [PMID: 36104214 PMCID: PMC10087978 DOI: 10.1002/ptr.7610] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/19/2023]
Abstract
Osteoporosis, a systemic bone disease that is characterized by a reduction in bone mass and destruction of bone microstructure, is becoming a serious problem worldwide. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into bone-forming osteoblasts, and play an important role in maintaining homeostasis of bone metabolism, thus being a potential therapeutic target for osteoporosis. Although the phytochemical alpinetin (APT) has been reported to possess a variety of pharmacological activities, it is still unclear whether APT can influence the osteogenic differentiation of on BMSCs and if it can improve osteoporosis. In this study, we found that APT treatment was able to enhance osteogenic differentiation levels of human BMSCs in vitro and mouse ones in vivo as revealed by multiple osteogenic markers including increased alkaline phosphatase activity and osteocalcin expression. Mechanistically, the protein kinase A (PKA)/mTOR/ULK1 signaling was involved in the action of APT to enhance the osteogenic differentiation of BMSCs. In addition, oral administration of APT significantly mitigated the bone loss in a dexamethasone-induced mouse model of osteoporosis through strengthening PKA signaling and autophagy. Altogether, these data demonstrate that APT promotes osteogenic differentiation in BMSCs by augmenting the PKA/mTOR/ULK1 autophagy signaling, highlighting its potential therapeutic application for treating osteoporotic diseases.
Collapse
Affiliation(s)
- Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Jinteng Li
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Mengjun Ma
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, People's Republic of China
| |
Collapse
|
22
|
He F, Yang C, Liu H, Wang J. Changes in the mechanical properties of human mesenchymal stem cells during differentiation. ROYAL SOCIETY OPEN SCIENCE 2023; 10:220607. [PMID: 36636310 PMCID: PMC9810430 DOI: 10.1098/rsos.220607] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 12/06/2022] [Indexed: 06/17/2023]
Abstract
A thorough understanding of the changes in mechanical property behind intracellular biophysical and biochemical processes during differentiation of human mesenchymal stem cells (hMSCs) is helpful to direct and enhance the commitment of cells to a particular lineage. In this study, displacement creep of the mesenchymal cell lineages (osteogenic, chondrogenic and adipogenic hMSCs) were determined by using atomic force microscopy, which was then used to determine their mechanical properties. We found that at any stages of differentiation, the mesenchymal cell lineages are linear viscoelastic materials and well matched with a simple power-law creep compliance. In addition, the viscoelasticity of mesenchymal cell lineages showed different trends during differentiation. The adipogenic hMSCs showed continuous softening at all stages. The osteogenic and chondrogenic hMSCs only continuously soften and become more fluid-like in the early stage of differentiation, and get stiffened and less fluid-like in the later stage. These findings will help more accurately imitate cellular biomechanics in the microenvironment, and provided an important reference in the biophysics biomimetic design of stem cell differentiation.
Collapse
Affiliation(s)
- Fei He
- Key Laboratory of Mechanics on Disaster and Environment in Western China, Ministry of Education, College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Chendong Yang
- Key Laboratory of Mechanics on Disaster and Environment in Western China, Ministry of Education, College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Haoye Liu
- Key Laboratory of Mechanics on Disaster and Environment in Western China, Ministry of Education, College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jizeng Wang
- Key Laboratory of Mechanics on Disaster and Environment in Western China, Ministry of Education, College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| |
Collapse
|
23
|
Bone Tissue and the Nervous System: What Do They Have in Common? Cells 2022; 12:cells12010051. [PMID: 36611845 PMCID: PMC9818711 DOI: 10.3390/cells12010051] [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: 09/29/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022] Open
Abstract
Degenerative diseases affecting bone tissues and the brain represent important problems with high socio-economic impact. Certain bone diseases, such as osteoporosis, are considered risk factors for the progression of neurological disorders. Often, patients with neurodegenerative diseases have bone fractures or reduced mobility linked to osteoarthritis. The bone is a dynamic tissue involved not only in movement but also in the maintenance of mineral metabolism. Bone is also associated with the generation of both hematopoietic stem cells (HSCs), and thus the generation of the immune system, and mesenchymal stem cells (MSCs). Bone marrow is a lymphoid organ and contains MSCs and HSCs, both of which are involved in brain health via the production of cytokines with endocrine functions. Hence, it seems clear that bone is involved in the regulation of the neuronal system and vice versa. This review summarizes the recent knowledge on the interactions between the nervous system and bone and highlights the importance of the interaction between nerve and bone cells. In addition, experimental models that study the interaction between nerve and skeletal cells are discussed, and innovative models are suggested to better evaluate the molecular interactions between these two cell types.
Collapse
|
24
|
Yang Y, Yuan L, Cao H, Guo J, Zhou X, Zeng Z. Application and Molecular Mechanisms of Extracellular Vesicles Derived from Mesenchymal Stem Cells in Osteoporosis. Curr Issues Mol Biol 2022; 44:6346-6367. [PMID: 36547094 PMCID: PMC9776574 DOI: 10.3390/cimb44120433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Osteoporosis (OP) is a chronic bone disease characterized by decreased bone mass, destroyed bone microstructure, and increased bone fragility. Accumulative evidence shows that extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) (MSC-EVs), especially exosomes (Exos), exhibit great potential in the treatment of OP. However, the research on MSC-EVs in the treatment of OP is still in the initial stage. The potential mechanism has not been fully clarified. Therefore, by reviewing the relevant literature of MSC-EVs and OP in recent years, we summarized the latest application of bone targeted MSC-EVs in the treatment of OP and further elaborated the potential mechanism of MSC-EVs in regulating bone formation, bone resorption, bone angiogenesis, and immune regulation through internal bioactive molecules to alleviate OP, providing a theoretical basis for the related research of MSC-EVs in the treatment of OP.
Collapse
Affiliation(s)
- Yajing Yang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Lei Yuan
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Hong Cao
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
- Correspondence: (X.Z.); (Z.Z.)
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Correspondence: (X.Z.); (Z.Z.)
| |
Collapse
|
25
|
Natesan K, Srivalli T, Mohan H, Jayaprakash A, Ramalingam V. UPLC-ESI-Q-TOF-MS E-based metabolomics analysis of Acer mono sap and evaluation of osteogenic activity in mouse osteoblast cells. Food Funct 2022; 13:13002-13013. [PMID: 36449013 DOI: 10.1039/d2fo01948e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Investigation of phytochemicals and bioactive molecules is tremendously vital for the applications of new plant resources in chemistry, food, and medicine. In this study, the chemical profiling of sap of Acer mono (SAM), a Korean syrup known for its anti-osteoporosis effect, was performed using UPLC-ESI-Q-TOF-MSE analysis. A total of 23 compounds were identified based on the mass and fragmentation characteristics and most of the compounds have significant biomedical applications. The in vitro antioxidant assessment of SAM indicated excellent activity by scavenging DPPH and ABTS-free radicals and were found to be 23.35 mg mL-1 and 29.33 mg mL-1, respectively, as IC50 concentrations. As well, the in vitro proliferation effect of the SAM was assessed against mouse MC3T3-E1 cells, and the results showed that the SAM enhanced the proliferation of the cells, and 12.5 mg mL-1 and 25 mg mL-1 of SAM were selected for osteogenic differentiation. The morphological analysis clearly evidenced the SAM enhanced the osteogenic activity in MC3T3-E1 cells by the increased deposition of extracellular calcium and nodule formation. Moreover, the qRT-PCR analysis confirmed the increased expression of osteoblast marker gene expression including ALP, osteocalcin, osteopontin, collagen1α1, Runx2, and osterix in SAM-treated MC3T3-E1 cells. Together, these results suggest that SAM possesses osteogenic effects and can be used for bone regeneration and bone loss-associated diseases such as osteoporosis.
Collapse
Affiliation(s)
- Karthi Natesan
- School of Allied Health Sciences, REVA University, Bengaluru, India
| | - Thimmarayan Srivalli
- PG and Research Department of Biochemistry, Scared Heart College (Autonomous), Tirupattur - 635601, Tamil Nadu, India (Affiliated to Thiruvalluvar University, Serkkadu, Vellore - 632115, Tamil Nadu, India)
| | - Harshavardhan Mohan
- Department of Chemistry, Research Institute of Physics and Chemistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Arul Jayaprakash
- PG and Research Department of Biochemistry, Scared Heart College (Autonomous), Tirupattur - 635601, Tamil Nadu, India (Affiliated to Thiruvalluvar University, Serkkadu, Vellore - 632115, Tamil Nadu, India)
| | - Vaikundamoorthy Ramalingam
- Centre for Natural Products & Traditional Knowledge, CSIR-Indian Institute of Chemical Technology, Hyderabad, India. .,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad - 201002, India
| |
Collapse
|
26
|
The Evaluation of Xiaozeng Qianggu Tablets for Treating Postmenopausal Osteoporosis via up-Regulated Autophagy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3960834. [PMID: 36193128 PMCID: PMC9526660 DOI: 10.1155/2022/3960834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022]
Abstract
Objective. Postmenopausal osteoporosis (PMOP) is a common age-associated disease in the life course. Clinically, Xiaozeng Qianggu Tablets (XQT) have a potent therapeutic effect on the PMOP. However, the bioactive components and the mechanism of XQT underlying the PMOP treatment were unclear and it should be explored to discover the scientific connotation in traditional medical practice. Methods. The components in XQT were identified by UPLC-Q-TOF/MS. The animal model of PMOP was established by surgical ovariectomy in the female Sprague-Dawley rats. After treatment of XQT, the therapeutic effect was assessed by the determination of bone metabolism biomarkers in serum and histopathological examination. The effect of XQT on the autophagy and bone micro-situation were tested using western blot, RT-qPCR, and transmission electron microscope. Results. There were 27 compounds identified in XQT, including catalpol, monotropein, verbascoside, cryptochlorogenic acid, 5,7-dihydroxychromone 7-rutinoside, biorobin, and so on. The bone metabolism markers (alkaline phosphatase, bone alkaline phosphatase, procollagen type I intact N-terminal propeptide, cross-linked carboxy-terminal telopeptide of type I collagen, and tartrate-resistant acid phosphatase) were significantly increased in the PMOP rats and reversed by XQT administration. Moreover, the width of bone trabeculae and the ratio of the area of calcium deposition to bone trabeculae were also improved after treating the middle dose of XQT. Meanwhile, the bone micro-structure was improved by XQT. The mRNA and protein expression of unc-51 like kinase 1, beclin-1, and microtubule-associated protein 1B-light chain 3 in PMOP rats were down-regulated and up-regulated by XQT administration. Conclusions. The compounds in XQT, including catalpol, monotropein, verbascoside cryptochlorogenic acid, and so on, were valuable for further pharmacy evaluation. The pathological changes and bone micro-structure were improved by XQT, and the down-regulated autophagy level was also restored, which suggested a potent effect of XQT on treating PMOP, corresponding to its clinic use.
Collapse
|
27
|
Zhang X, He J, Qiao L, Wang Z, Zheng Q, Xiong C, Yang H, Li K, Lu C, Li S, Chen H, Hu X. 3D
printed
PCLA
scaffold with nano‐hydroxyapatite coating doped green tea
EGCG
promotes bone growth and inhibits multidrug‐resistant bacteria colonization. Cell Prolif 2022; 55:e13289. [PMID: 35791492 PMCID: PMC9528762 DOI: 10.1111/cpr.13289] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023] Open
Affiliation(s)
- Xiangchun Zhang
- Tea Research Institute, Chinese Academy of Agricultural Sciences Hangzhou China
| | - Jian He
- College of Medical, Henan University of Science and Technology Luoyang China
| | - Liang Qiao
- The First Affiliated Hospital College of Clinical Medicine of Henan University of Science and Technology Luoyang People's Republic of China
| | - Ziqi Wang
- Tea Research Institute, Chinese Academy of Agricultural Sciences Hangzhou China
| | - Qinqin Zheng
- Tea Research Institute, Chinese Academy of Agricultural Sciences Hangzhou China
| | - Chengdong Xiong
- Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences Chengdu Sichuan China
| | - Hui Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and West China Hospital of Stomatology Sichuan University Chengdu China
| | - Kainan Li
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University Chengdu China
| | - Chengyin Lu
- Tea Research Institute, Chinese Academy of Agricultural Sciences Hangzhou China
| | - Sanqiang Li
- College of Medical, Henan University of Science and Technology Luoyang China
| | - Hongping Chen
- Tea Research Institute, Chinese Academy of Agricultural Sciences Hangzhou China
| | - Xulin Hu
- Clinical Medical College and Affiliated Hospital of Chengdu University, Chengdu University Chengdu China
| |
Collapse
|
28
|
Guo M, Liu F, Wang W, Liu Z, Zhu Z, Liu Y, Huang Z. Naringin Promotes Osteogenic/Odontogenic Differentiation of Dental Pulp Stem Cells via Wnt/ β-Catenin. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:4505471. [PMID: 35677363 PMCID: PMC9168102 DOI: 10.1155/2022/4505471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022]
Abstract
Purpose This investigation intended to unravel the effect and mechanism of naringin on the proliferation and osteogenic differentiation of human dental pulp stem cells (hDPSCs). Methods hDPSCs were induced to differentiate, and the degree of cell differentiation was observed by alizarin red staining, Oil Red O staining, and Alcian blue staining. hDPSCs were treated with 0, 20, 40, and 80 μmol/L naringin for 48 h, respectively. The proliferation rate and chemotaxis of the cells were measured by MTT and transwell assay, alkaline phosphatase (ALP) activity and osteogenic differentiation degree by ALP staining and alizarin red staining, and gene expression of osteogenic markers by qRT-PCR. Additionally, western blot was performed to test the levels of Wnt/β-catenin signaling-related proteins in hDPSCs. Results The isolated hDPSCs with spindle-shaped morphology had good differentiation capability. Further experiments confirmed naringin-caused increases in the proliferation rate and migration ability of hDPSCs. In addition, compared with the control group, naringin-treated cells had strong ALP activity and ossification levels and higher expression of Runx2, OPN, DSPP, and DMP1. The western blot results showed that naringin significantly activated Wnt/β-catenin signaling in hDPSCs. Conclusion Taken together, naringin enhances the proliferation, migration, and osteogenesis of hDPSCs through stimulating Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Meiling Guo
- Department of General Dentistry, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
| | - Fen Liu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Wenjuan Wang
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Zhirong Liu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Zhipeng Zhu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Yiyu Liu
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| | - Zhen Huang
- The Key Laboratory of Oral Biomedicine, Nanchang City, Jiangxi Province 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang City, Jiangxi Province 330006, China
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanchang University, Nanchang City, Jiangxi Province 330006, China
| |
Collapse
|
29
|
Ma Q, Song C, Yin B, Shi Y, Ye L. The role of Trithorax family regulating osteogenic and Chondrogenic differentiation in mesenchymal stem cells. Cell Prolif 2022; 55:e13233. [PMID: 35481717 PMCID: PMC9136489 DOI: 10.1111/cpr.13233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/17/2022] [Accepted: 03/28/2022] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) hold great promise and clinical efficacy in bone/cartilage regeneration. With a deeper understanding of stem cell biology over the past decade, epigenetics stands out as one of the most promising ways to control MSCs differentiation. Trithorax group (TrxG) proteins, including the COMPASS family, ASH1L, CBP/p300 as histone modifying factors, and the SWI/SNF complexes as chromatin remodelers, play an important role in gene expression regulation during the process of stem cell differentiation. This review summarises the components and functions of TrxG complexes. We provide an overview of the regulation mechanisms of TrxG in MSCs osteogenic and chondrogenic differentiation, and discuss the prospects of epigenetic regulation mediated by TrxG in bone and cartilage regeneration.
Collapse
Affiliation(s)
- Qingge Ma
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenghao Song
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bei Yin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Shi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Tang C, Liang D, Qiu Y, Zhu J, Tang G. Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis. Mol Med Rep 2022; 25:132. [PMID: 35179221 PMCID: PMC8867465 DOI: 10.3892/mmr.2022.12648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/13/2021] [Indexed: 11/25/2022] Open
Abstract
Osteoporosis is a bone-related disease that results from impaired bone formation and excessive bone resorption. The potential value of adipokines has been investigated previously, due to their influence on osteogenesis. However, the osteogenic effects induced by omentin-1 remain unclear. The aim of the present study was to determine the regulatory effects of omentin-1 on osteoblast viability and differentiation, as well as to explore the underlying molecular mechanism. The present study investigated the effects of omentin-1 on the viability and differentiation of mouse pre-osteoblast cells (MC3T3-E1) using quantitative and qualitative measures. A Cell Counting Kit-8 assay was used to assess the viability of MC3T3-E1 cells following treatment with different doses of omentin-1. Omentin-1 and bone morphogenetic protein (BMP) inhibitor were added to osteogenic induction mediums in different ways to assess their effect. The alkaline phosphatase (ALP) activity and Alizarin Red S (ARS) staining of MC3T3-E1 cells treated with omentin-1 and/or BMP inhibitor were used to examine the effects of omentin-1 on differentiation and mineralization. Western blotting was used to further explore its potential mechanism, and to study the role of omentin-1 on the viability and differentiation of osteoblasts. The results showed that omentin-1 altered the viability of MC3T3-E1 cells in a dose-dependent manner. Omentin-1 treatment significantly increased the expression of members of the TGF-β/Smad signaling pathway. In the omentin-1 group, the ALP activity of the MC3T3-E1 cells was increased, and the ARS staining area was also increased. The mRNA and protein expression levels of BMP2, Runt-related transcription factor 2, collagen1, osteopontin, osteocalcin and osterix in the omentin-1 group were also significantly upregulated. All these effects were reversed following treatment with SIS3 HCl. These results demonstrated that omentin-1 can significantly promote osteoblast viability and differentiation via the TGF-β/Smad signaling pathway, thereby promoting bone formation and preventing osteoporosis.
Collapse
Affiliation(s)
- Cuisong Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, Shanghai 200072, P.R. China
| | - Dengpan Liang
- Department of Cardiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yuyou Qiu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jingqi Zhu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guangyu Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, Shanghai 200072, P.R. China
| |
Collapse
|
31
|
Li H, Li M, Ran X, Cui J, Wei F, Yi G, Chen W, Luo X, Chen Z. The Role of Zinc in Bone Mesenchymal Stem Cell Differentiation. Cell Reprogram 2022; 24:80-94. [PMID: 35172118 DOI: 10.1089/cell.2021.0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Zinc is an essential trace element for bone growth and bone homeostasis in the human body. Bone mesenchymal stem cells (BMSCs) are multipotent progenitors existing in the bone marrow stroma with the capability of differentiating along multiple lineage pathways. Zinc plays a paramount role in BMSCs, which can be spurred differentiating into osteoblasts, chondrocytes, or adipocytes, and modulates the formation and activity of osteoclasts. The expression of related genes also changed during the differentiation of various cell phenotypes. Based on the important role of zinc in BMSC differentiation, using zinc as a therapeutic approach for bone remodeling will be a promising method. This review explores the role of zinc ion in the differentiation of BMSCs into various cell phenotypes and outlines the existing research on their molecular mechanism.
Collapse
Affiliation(s)
- Huiyun Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Muzhe Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xun Ran
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Juncheng Cui
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Fu Wei
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Guoliang Yi
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Wei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Xuling Luo
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhiwei Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
32
|
Felipetti FA, Costa VSLP, Neves JDS, Sousa IG, Piedade SMDS, Novaes PD. Látex de Hancornia speciosa Gomes aumenta a mineralização óssea em ratos: Um estudo pré-clínico. Rev Bras Ortop 2022; 57:488-495. [PMID: 35785126 PMCID: PMC9246514 DOI: 10.1055/s-0041-1741020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/09/2021] [Indexed: 10/27/2022] Open
Abstract
Resumo
Objetivo Avaliar o efeito sistêmico do látex de Hancornia especiosa na neoformação óssea e mineralização em ratos.
Métodos Para isso, primeiro o látex foi coletado, e sua composição foi analisada. No estudo, foram utilizados 30 ratos Wistar machos submetidos simultaneamente a dois procedimentos cirúrgicos: extração de incisivo e criação de um defeito de 2 mm de diâmetro no osso parietal. Os ratos foram divididos em dois grupos: controle sistêmico (CS) e látex sistêmico (XS), aos quais foi administrado, oral e diariamente, 1,5 mL de água ou uma solução contendo 50% de água e 50% de látex por gavagem, respectivamente. Após 15 dias do tratamento, os animais foram eutanizados, e suas amostras, coletadas.
Resultados Os resultados foram analisados estatisticamente, e o nível de significância foi fixado em 0,05. Mostramos que o látex de H. speciosa continha cálcio. A administração oral e diária deste látex por 15 dias aumentou o conteúdo de cálcio e fósforo de osso basal e de osso recém-formado no alvéolo mandibular de ratos.
Conclusão Este foi um estudo pioneiro, que demonstrou o potencial do látex de H. speciosa no aumento da mineralização óssea. Nossos resultados podem ajudar na concepção e no desenvolvimento de uma droga natural.
Collapse
Affiliation(s)
- Francielly Andressa Felipetti
- Departamento de Morfologia, Faculdade de Odontologia de Piracicaba (FOP), Universidade Estadual de Campinas (Unicamp), Piracicaba, SP, Brasil
| | | | - Juliana dos Santos Neves
- Departamento de Morfologia, Faculdade de Odontologia de Piracicaba (FOP), Universidade Estadual de Campinas (Unicamp), Piracicaba, SP, Brasil
| | - Ingrid Grazielle Sousa
- Departamento de Morfologia, Faculdade de Odontologia de Piracicaba (FOP), Universidade Estadual de Campinas (Unicamp), Piracicaba, SP, Brasil
| | - Sônia Maria De Stefano Piedade
- Departamento de Ciências Exatas, Escola Superior de Agricultura Luiz de Queiroz (ESALQ), Universidade de São Paulo (USP), Piracicaba, SP, Brasil
| | - Pedro Duarte Novaes
- Departamento de Morfologia, Faculdade de Odontologia de Piracicaba (FOP), Universidade Estadual de Campinas (Unicamp), Piracicaba, SP, Brasil
| |
Collapse
|
33
|
Liao HH, Livneh H, Chung YJ, Lin CH, Lai NS, Yen HR, Tsai TY. A Comparison of the Risk of Fracture in Rheumatoid Arthritis Patients with and without Receiving Chinese Herbal Medicine. J Multidiscip Healthc 2021; 14:3399-3409. [PMID: 34934326 PMCID: PMC8684401 DOI: 10.2147/jmdh.s334134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 12/12/2022] Open
Abstract
Objective Patients with rheumatoid arthritis (RA) often suffer from bone complications due to persistent joint inflammation, especially incident fracture. Nowadays, Chinese herbal medicines (CHMs) have provided safe and effective therapy for treating skeletal conditions, but it is unclear whether CHMs can prevent fracture onset among RA individuals. This study aimed to determine the association between the use of CHMs and the risk of fracture among them. Methods This retrospective, population-based study retrieved administrative health data from the Taiwan National Health Insurance (NHI) database to identify patients with newly diagnosed RA between 2000 and 2009. Of the 6178 incident RA patients, 2495 matched pairs of CHMs users and non-CHMs users were identified by propensity score matching. Enrollees with hip fractures prior to RA onset were excluded. Included subjects were followed until the end of 2013. Incidence and adjusted hazard ratios (HR) of new-onset bone fracture in the multivariable Cox proportional hazard model were measured with 95% confidence interval (CI). Results Fracture incidence was lower in CHMs users than in the comparison cohort (26.91 vs 32.94 per 1000 person-years, respectively), with an adjusted HR of 0.82 (95% CI: 0.73-0.92). Subjects receiving CHMs for more than 2 years had a much lower risk of fracture onset by more than 50%. Some CHMs prescriptions (Yan Hu Suo, Bei Mu, Da Huang, Dang Shen, Fu-Zi, Shu-Jing-Huo-Xue-Tang, Dang-Gui-Nian-Tong-Tang, Jia-Wei-Xiao-Yao-San, Gan-Lu-Yin, and Gui-Zhi-Shao-Yao-Zhi-Mu-Tang) were associated with reduced fracture risk. Conclusion Adding CHMs to routine treatment was found to be related to lower fracture risk in RA patients.
Collapse
Affiliation(s)
- Hou-Hsun Liao
- Department of Chinese Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Dalin Township, Chiayi, 62247, Taiwan.,Graduate Institute of Chinese Medicine, School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, Tzu Chi University of Science and Technology, Hualien, 62247, Taiwan
| | - Hanoch Livneh
- Rehabilitation Counseling Program, Portland State University, Portland, OR, 97207-0751, USA
| | - Yu-Jung Chung
- Department of Chinese Medicine, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Dalin Township, Chiayi, 62247, Taiwan
| | - Ching-Hsing Lin
- Department of Orthopedics, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan.,School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Ning-Sheng Lai
- School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan.,Division of Allergy, Immunology and Rheumatology, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
| | - Hung-Rong Yen
- Graduate Institute of Chinese Medicine, School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Chinese Medicine, China Medical University Hospital, Taichung, 404, Taiwan.,Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, 404, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, 404, Taiwan.,Department of Biotechnology, Asia University, Taichung, 413, Taiwan
| | - Tzung-Yi Tsai
- Department of Nursing, Tzu Chi University of Science and Technology, Hualien, 62247, Taiwan.,Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, 70428, Taiwan.,Department of Medical Research, Dalin Tzu Chi Hospital, The Buddhist Tzu Chi Medical Foundation, Chiayi, 62247, Taiwan
| |
Collapse
|
34
|
Teixeira S, Santos MM, Branco LC, Costa-Rodrigues J. Etidronate-based organic salts and ionic liquids: In vitro effects on bone metabolism. Int J Pharm 2021; 610:121262. [PMID: 34748807 DOI: 10.1016/j.ijpharm.2021.121262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 11/26/2022]
Abstract
Bisphosphonates are a class of drugs widely used for the treatment of several pathologies associated with increased bone resorption. Although displaying low oral bioavailability, these drugs have the ability to accumulate in bone matrix, where the biological effects are exerted. In the present work, four mono- and dianionic Etidronate-based Organic Salts and Ionic Liquids (Eti-OSILs) were developed by combination of this drug with the superbases 1,1,3,3-tetramethylguanidine (TMG) and 1,5-diazabicyclo(4.3.0)non-5-ene (DBN) as cations, aiming to improve not only the physicochemical properties of this seminal bisphosphonate, but also its efficacy in the modulation of cellular behavior, particularly on human osteoclasts and osteoblasts. It was observed that some of the developed compounds, in particular the dianionic ones, presented very high water solubility and diminished or absent polymorphism. Also, several of them appeared to be more cytotoxic against human breast and osteosarcoma cancer cell lines while retaining low toxicity to normal cells. Regarding bone cells, a promotion of an anabolic state was observed for all Eti-OSILs, primarily for the dianionic ones, which leads to an inhibition of osteoclastogenesis and an increase in osteoblastogenesis. The observed effects resulted from differential modulation of intracellular signaling pathways by the Eti-OSILs in comparison with Etidronate. Hence, these results pave the way for the development of more efficient and bioavailable ionic formulations of bisphosphonates aiming to effectively modulate bone metabolism, particularly in the case of increased bone resorption.
Collapse
Affiliation(s)
- Sónia Teixeira
- Instituto de Ciências Biomédicas Abel Salazar, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| | - Miguel M Santos
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| | - Luís C Branco
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal.
| | - João Costa-Rodrigues
- ESS - Escola Superior de Saúde, Politécnico do Porto, R. Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal; Instituto Politécnico de Viana do Castelo, Escola Superior de Saúde, Rua D. Moisés Alves Pinho 190, 4900-314 Viana do Castelo, Portugal; i3S, Instituto de Inovação e Investigação em Saúde, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal.
| |
Collapse
|
35
|
FGF19 protects against obesity-induced bone loss by promoting osteogenic differentiation. Biomed Pharmacother 2021; 146:112524. [PMID: 34906775 DOI: 10.1016/j.biopha.2021.112524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/20/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Human fibroblast growth factor 19 (FGF19) has become a potential therapeutic target for metabolic-related diseases. However, the effects of FGF19 on obesity-induced bone loss have not been completely elucidated. The aim of this study was to investigate the protective effects of FGF19 in high-fat diet (HFD)-fed obese mice and palmitic acid (PA)-treated osteoblasts and to further explore its underlying mechanisms. In vivo, we found that FGF19 alleviated the decreased bone mineral density (BMD) induced by HFD. Micro-CT analysis of femur samples and histological analysis indicated that FGF19 alleviated HFD-induced loss of bone trabeculae and damage to the bone trabecular structure. In vitro, the results suggested that FGF19 ameliorated the PA-induced decline in osteoblast proliferation, increased cell death and impaired cell morphology. Additionally, FGF19 protected against the decline in activation of alkaline phosphatase (ALP) and protein expression of Collagen-1, Runx-2, and osteopontin (OPN) induced by PA. Furthermore, FGF19 might enhance osteogenic differentiation via the Wnt/β-catenin pathway and inhibit osteoclastogenesis by regulating the osteoprotegerin (OPG)/receptor activator of NF-κB ligand (RANKL) axis, thus attenuating the negative effect of PA in osteoblasts. In conclusion, our results suggested that FGF19 might promote osteogenic differentiation partially through activation of the Wnt/β-catenin pathway and alleviate obesity-induced bone loss.
Collapse
|
36
|
Wang Z, Chai C, Wang R, Feng Y, Huang L, Zhang Y, Xiao X, Yang S, Zhang Y, Zhang X. Single-cell transcriptome atlas of human mesenchymal stem cells exploring cellular heterogeneity. Clin Transl Med 2021; 11:e650. [PMID: 34965030 PMCID: PMC8715893 DOI: 10.1002/ctm2.650] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/24/2021] [Accepted: 10/30/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The heterogeneity of mesenchymal stem cells (MSCs) is poorly understood, thus limiting clinical application and basic research reproducibility. Advanced single-cell RNA sequencing (scRNA-seq) is a robust tool used to analyse for dissecting cellular heterogeneity. However, the comprehensive single-cell atlas for human MSCs has not been achieved. METHODS This study used massive parallel multiplexing scRNA-seq to construct an atlas of > 130 000 single-MSC transcriptomes across multiple tissues and donors to assess their heterogeneity. The most widely clinically utilised tissue resources for MSCs were collected, including normal bone marrow (n = 3), adipose (n = 3), umbilical cord (n = 2), and dermis (n = 3). RESULTS Seven tissue-specific and five conserved MSC subpopulations with distinct gene-expression signatures were identified from multiple tissue origins based on the high-quality data, which has not been achieved previously. This study showed that extracellular matrix (ECM) highly contributes to MSC heterogeneity. Notably, tissue-specific MSC subpopulations were substantially heterogeneous on ECM-associated immune regulation, antigen processing/presentation, and senescence, thus promoting inter-donor and intra-tissue heterogeneity. The variable dynamics of ECM-associated genes had discrete trajectory patterns across multiple tissues. Additionally, the conserved and tissue-specific transcriptomic-regulons and protein-protein interactions were identified, potentially representing common or tissue-specific MSC functional roles. Furthermore, the umbilical-cord-specific subpopulation possessed advantages in immunosuppressive properties. CONCLUSION In summary, this work provides timely and great insights into MSC heterogeneity at multiple levels. This MSC atlas taxonomy also provides a comprehensive understanding of cellular heterogeneity, thus revealing the potential improvements in MSC-based therapeutic efficacy.
Collapse
Affiliation(s)
- Zheng Wang
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
| | - Chengyan Chai
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
| | - Rui Wang
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
| | - Yimei Feng
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
| | - Lei Huang
- Department of Urologythe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Yiming Zhang
- Department of Plastic and Cosmetic Surgerythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
| | - Xia Xiao
- Time Plastic Surgery HospitalChongqingChina
| | - Shijie Yang
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
| | - Yunfang Zhang
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
| | - Xi Zhang
- Medical Center of Hematologythe Second Affiliated HospitalArmy Medical UniversityChongqingChina
- State Key Laboratory of TraumaBurn and Combined InjuryArmy Medical UniversityChongqingChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
37
|
Zhang C, Zhu H, Ren X, Gao B, Cheng B, Liu S, Sha B, Li Z, Zhang Z, Lv Y, Wang H, Guo H, Lu TJ, Xu F, Genin GM, Lin M. Mechanics-driven nuclear localization of YAP can be reversed by N-cadherin ligation in mesenchymal stem cells. Nat Commun 2021; 12:6229. [PMID: 34711824 PMCID: PMC8553821 DOI: 10.1038/s41467-021-26454-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 10/01/2021] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cells adopt differentiation pathways based upon cumulative effects of mechanosensing. A cell's mechanical microenvironment changes substantially over the course of development, beginning from the early stages in which cells are typically surrounded by other cells and continuing through later stages in which cells are typically surrounded by extracellular matrix. How cells erase the memory of some of these mechanical microenvironments while locking in memory of others is unknown. Here, we develop a material and culture system for modifying and measuring the degree to which cells retain cumulative effects of mechanosensing. Using this system, we discover that effects of the RGD adhesive motif of fibronectin (representative of extracellular matrix), known to impart what is often termed "mechanical memory" in mesenchymal stem cells via nuclear YAP localization, are erased by the HAVDI adhesive motif of the N-cadherin (representative of cell-cell contacts). These effects can be explained by a motor clutch model that relates cellular traction force, nuclear deformation, and resulting nuclear YAP re-localization. Results demonstrate that controlled storage and removal of proteins associated with mechanical memory in mesenchymal stem cells is possible through defined and programmable material systems.
Collapse
Affiliation(s)
- Cheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Hongyuan Zhu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Xinru Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Bin Gao
- Department of Endocrinology, Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, People's Republic of China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shaobao Liu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, People's Republic of China
| | - Baoyong Sha
- School of Basic Medical Science, Xi'an Medical University, Xi'an, 710021, People's Republic of China
| | - Zhaoqing Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Zheng Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, People's Republic of China
| | - Haohua Wang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, People's Republic of China
| | - Hui Guo
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, People's Republic of China
- MOE Key Laboratory of Multifunctional Materials and Structures, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, 63130, MO, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, 63130, MO, USA
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| |
Collapse
|
38
|
Tevlek A, Aydin HM. Multi-layered in vitro 3D-bone model via combination of osteogenic cell sheets with electrospun membrane interlayer. J Biomater Appl 2021; 36:818-833. [PMID: 34162235 DOI: 10.1177/08853282211027889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, it was aimed to present an approach for the development of multi-layered tissue engineering constructs by using cell sheet engineering. Briefly, MC3T3-E1 mouse pre-osteoblast cells were cultured in temperature-responsive plates (Nunc Upcell®) in the presence of osteogenic medium and the resulting cell sheets were laminated with electrospun poly(L-lactic acid) (PLLA) membranes to obtain viable three-dimensional, thick constructs. The constructs prepared without PLLA membranes were used as control. The cell viability and death in the resulting structures were investigated by microscopic and colorimetric methods. The in vitro performance of the structures was discussed comparatively. Alkaline phosphatase (ALP) activity, collagen and sulfated glycosaminoglycan (sGAG) content values were calculated. The presented approach shows potential for engineering applications of complex tissues with at least two or more microenvironments such as osteochondral, corneal or vascular tissues.
Collapse
Affiliation(s)
- Atakan Tevlek
- Institute of Science, Hacettepe University, Ankara, Turkey
| | - Halil Murat Aydin
- Institute of Science, Hacettepe University, Ankara, Turkey.,Centre for Bioengineering, Hacettepe University, Ankara, Turkey
| |
Collapse
|
39
|
Donsante S, Palmisano B, Serafini M, Robey PG, Corsi A, Riminucci M. From Stem Cells to Bone-Forming Cells. Int J Mol Sci 2021; 22:ijms22083989. [PMID: 33924333 PMCID: PMC8070464 DOI: 10.3390/ijms22083989] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/06/2021] [Accepted: 04/10/2021] [Indexed: 12/22/2022] Open
Abstract
Bone formation starts near the end of the embryonic stage of development and continues throughout life during bone modeling and growth, remodeling, and when needed, regeneration. Bone-forming cells, traditionally termed osteoblasts, produce, assemble, and control the mineralization of the type I collagen-enriched bone matrix while participating in the regulation of other cell processes, such as osteoclastogenesis, and metabolic activities, such as phosphate homeostasis. Osteoblasts are generated by different cohorts of skeletal stem cells that arise from different embryonic specifications, which operate in the pre-natal and/or adult skeleton under the control of multiple regulators. In this review, we briefly define the cellular identity and function of osteoblasts and discuss the main populations of osteoprogenitor cells identified to date. We also provide examples of long-known and recently recognized regulatory pathways and mechanisms involved in the specification of the osteogenic lineage, as assessed by studies on mice models and human genetic skeletal diseases.
Collapse
Affiliation(s)
- Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Marta Serafini
- Centro Ricerca M. Tettamanti, Clinica Pediatrica, Università di Milano-Bicocca, Ospedale San Gerardo, 20900 Monza, Italy;
| | - Pamela G. Robey
- Skeletal Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA;
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina 324, 00161 Rome, Italy; (S.D.); (B.P.); (A.C.)
- Correspondence:
| |
Collapse
|
40
|
Xie J, Lou Q, Zeng Y, Liang Y, Xie S, Xu Q, Yuan L, Wang J, Jiang L, Mou L, Lin D, Zhao M. Single-Cell Atlas Reveals Fatty Acid Metabolites Regulate the Functional Heterogeneity of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:653308. [PMID: 33912565 PMCID: PMC8075002 DOI: 10.3389/fcell.2021.653308] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/09/2021] [Indexed: 12/28/2022] Open
Abstract
Bone marrow mesenchymal stem cells (MSCs) are widely used clinically due to their versatile roles in multipotency, immunomodulation, and hematopoietic stem cell (HSC) niche function. However, cellular heterogeneity limits MSCs in the consistency and efficacy of their clinical applications. Metabolism regulates stem cell function and fate decision; however, how metabolites regulate the functional heterogeneity of MSCs remains elusive. Here, using single-cell RNA sequencing, we discovered that fatty acid pathways are involved in the regulation of lineage commitment and functional heterogeneity of MSCs. Functional assays showed that a fatty acid metabolite, butyrate, suppressed the self-renewal, adipogenesis, and osteogenesis differentiation potential of MSCs with increased apoptosis. Conversely, butyrate supplement significantly promoted HSC niche factor expression in MSCs, which suggests that butyrate supplement may provide a therapeutic approach to enhance their HSC niche function. Overall, our work demonstrates that metabolites are essential to regulate the functional heterogeneity of MSCs.
Collapse
Affiliation(s)
- Jiayi Xie
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qi Lou
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China.,The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Yunxin Zeng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yingying Liang
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China.,The First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People's Hospital, Shenzhen, China
| | - Siyu Xie
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Quanhui Xu
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Lisha Yuan
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Jin Wang
- Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Linjia Jiang
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lisha Mou
- Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Meng Zhao
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Shenzhen Lansi Institute of Artificial Intelligence in Medicine, Shenzhen, China.,Key Laboratory of Stem Cells and Tissue Engineering, Zhongshan School of Medicine, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| |
Collapse
|
41
|
Shim NY, Ryu JI, Heo JS. Osteoinductive function of fucoidan on periodontal ligament stem cells: Role of PI3K/Akt and Wnt/β-catenin signaling pathways. Oral Dis 2021; 28:1628-1639. [PMID: 33682270 DOI: 10.1111/odi.13829] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/02/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES Fucoidan has been focused as a multifunctional therapeutic uses including bone health supplements. However, the critical molecular mechanisms of fucoidan for bone therapeutic agents have not been fully understood. We investigated the osteoinductive effect of fucoidan on periodontal ligament stem cells (PDLSCs) and how this polymer encouraged PDLSC osteogenesis. MATERIALS AND METHODS Osteogenic induction of PDLSCs was processed by culturing cells with fucoidan treatment. Osteogenic differentiation of PDLSCs was verified by alkaline phosphatase (ALP) activity, matrix mineralization assay, intracellular calcium levels, and mRNA expression and protein levels of osteogenic markers. RESULTS Fucoidan treatment showed higher osteogenic activity in the PDLSCs than the control groups. PDLSCs with fucoidan also presented increased levels of the phosphatidylinositol-3-kinase (PI3K) isoforms, p110α and p110γ compared to control cells. The phosphorylation of Akt, a PI3K downstream effector, was significantly increased at 90 min of fucoidan induction. Expression of β-catenin, a coactivator of canonical Wnt pathways, was increased in PDLSCs with fucoidan. β-catenin was found to link with PI3K activation during the fucoidan stimulation. When cells were blocked by PI3K inhibitor or β-catenin-specific siRNA, fucoidan-induced osteogenic activity of PDLSCs was significantly attenuated. CONCLUSION These findings suggest that the fucoidan stimulates osteogenic differentiation of PDLSCs via the PI3K/Akt and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Na Young Shim
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, Seoul, South Korea
| | - Jae-In Ryu
- Department of Preventive and Social Dentistry, School of Dentistry, Kyung Hee University, Seoul, South Korea
| | - Jung Sun Heo
- Department of Maxillofacial Biomedical Engineering and Institute of Oral Biology, School of Dentistry, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
42
|
Deng Y, Zhu W, Anhua Lin, Wang C, Xiong C, Xu F, Li J, Huang S, Zhang N, Huo Y. Exendin-4 promotes bone formation in diabetic states via HDAC1-Wnt/β-catenin axis. Biochem Biophys Res Commun 2021; 544:8-14. [PMID: 33516884 DOI: 10.1016/j.bbrc.2021.01.039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 01/13/2021] [Indexed: 02/08/2023]
Abstract
Exendin-4 has been found to have hypoglycemic effect and prevent bone loss in diabetic patients, but its mechanism of preventing bone loss is still unclear. In this study, high-fat diet combined with streptozotocin was used to establish type 2 diabetes mellitus (T2DM) mice, and bone marrow mesenchyme stem cells (BMSCs) were isolated for osteogenic induction in vitro. Alizarin red staining and ALP activity detection were used to observe the effect of exendin-4 on osteogenic differentiation of BMSCs. Western blot was used to detect the proteins expression in BMSCs. In vivo, the effects of exendin-4 treatment on body weight, blood glucose, bone density and bone quality of T2DM mice were observed by treatment with exendin-4. The results showed that exendin-4 promoted osteogenic differentiation of T2DM derived BMSCs, down-regulated histone deacetylase 1 (HDAC1) and p-β-Catenin proteins expression, and up-regulated Wnt3, β-Catenin and runt-related transcription factor 2 (Runx 2) proteins expression. In vivo, exendin-4 effectively suppressed the blood glucose and increased body weight of T2DM mice, and significantly improved bone density and bone quality of the right tibia. Interestingly, by over-expression of HDAC1 in BMSCs, the effect of exendin-4 on promoting osteogenic differentiation of BMSCs was attenuated, and the regulation of Wnt3a, β-Catenin, p-β-Catenin or Runx2 proteins were reversed. By injecting adenovirus containing HDAC1 into the right tibia of mice, the effect of exendin-4 on bone density and bone quality of T2DM mice was significantly attenuated. All above results suggest that the HDAC1-Wnt/β-Catenin signal axis is involved in the anti-diabetic bone loss effect of exendin-4, and HDAC1 may be the target of exendin-4.
Collapse
Affiliation(s)
- Ying Deng
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Wenyi Zhu
- Medical Department of Graduate School, Nanchang University, Nanchang, PR China
| | - Anhua Lin
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Chenxiu Wang
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Changhui Xiong
- Department of Science and Education, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Fanghua Xu
- Pathology Department, Pingxiang People's Hospital of Southern Medical University, Pingxiang, Jiangxi, 337055, PR China
| | - Jinfeng Li
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Shuijin Huang
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Na Zhang
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Yanan Huo
- Endocrinology Department, Jiangxi Provincial People(')s Hospital Affiliated to Nanchang University, Nanchang, Jiangxi, 330006, PR China.
| |
Collapse
|
43
|
Smith CO, Eliseev RA. Energy Metabolism During Osteogenic Differentiation: The Role of Akt. Stem Cells Dev 2021; 30:149-162. [PMID: 33307974 DOI: 10.1089/scd.2020.0141] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Osteogenic differentiation, the process by which bone marrow mesenchymal stem/stromal (a.k.a. skeletal stem) cells and osteoprogenitors form osteoblasts, is a critical event for bone formation during development, fracture repair, and tissue maintenance. Extra cellular and intracellular signaling pathways triggering osteogenic differentiation are relatively well known; however, the ensuing change in cell energy metabolism is less clearly defined. We and others have previously reported activation of mitochondria during osteogenic differentiation. To further elucidate the involved bioenergetic mechanisms and triggers, we tested the effect of osteogenic media containing ascorbate and β-glycerol phosphate, or various osteogenic hormones and growth factors on energy metabolism in long bone (ST2)- and calvarial bone (MC3T3-E1)-derived osteoprogenitors. We show that osteogenic media and differentiation factors, Wnt3a and BMP2, stimulate mitochondrial oxidative phosphorylation (OxPhos) with little effect on glycolysis. The activation of OxPhos occurs acutely, suggesting a metabolic signaling change rather than protein expression change. To this end, we found that the observed mitochondrial activation is Akt dependent. Akt is activated by osteogenic media, Wnt3a, and BMP2, leading to increased phosphorylation of various mitochondrial Akt targets, a phenomenon known to stimulate OxPhos. In sum, our data provide comprehensive analysis of cellular bioenergetics during osteoinduction in cells of two different origins (mesenchyme vs neural crest) and identify Wnt3a and BMP2 as physiological stimulators of mitochondrial respiration through Akt activation.
Collapse
Affiliation(s)
- Charles Owen Smith
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, Rochester, New York, USA
| | - Roman A Eliseev
- Center for Musculoskeletal Research, University of Rochester School of Medicine & Dentistry, Rochester, New York, USA
| |
Collapse
|
44
|
Li Y, Wang J, Ma Y, Du W, Feng H, Feng K, Li G, Wang S. MicroRNA-15b shuttled by bone marrow mesenchymal stem cell-derived extracellular vesicles binds to WWP1 and promotes osteogenic differentiation. Arthritis Res Ther 2020; 22:269. [PMID: 33198785 PMCID: PMC7667798 DOI: 10.1186/s13075-020-02316-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/11/2020] [Indexed: 01/08/2023] Open
Abstract
Background Osteogenic differentiation is an essential process for bone regeneration involving bone marrow mesenchymal stem cells (BMSCs). BMSC-secreted extracellular vesicles (EVs) enriched with microRNAs (miRs) have vital roles to play in mediating osteogenic differentiation. Therefore, this study aimed to explore the effect of BMSC-derived EVs loaded with miR-15b on osteogenic differentiation. Methods Human BMSCs (hBMSCs) were cultured and treated with plasmids overexpressing or knocking down KLF2, WWP1, and miR-15b to define the role of derived EVs in osteogenic differentiation in vitro. The expression of osteogenic differentiation-related marker was measured by Western blot analysis. The interaction among miR-15b, WWP1, and ubiquitination of KLF2 was investigated by dual-luciferase reporter, immunoprecipitation, and GST pull-down assays. Moreover, EVs from hBMSCs transfected with miR-15b inhibitor (EV-miR-15b inhibitor) were injected into ovariectomized rats to verify the effect of miR-15b on bone loss in vivo. Results WWP1 was downregulated, and KLF2 was upregulated during osteogenic differentiation. After co-culture with EVs, miR-15b expression was elevated and WWP1 expression was reduced in hBMSCs. Upregulation of miR-15b or KLF2 or downregulation of WWP1 or NF-κB increased ALP activity and cell mineralization, as well as osteogenic differentiation-related marker expression in hBMSCs. Mechanistically, miR-15b targeted and inhibited WWP1, thus attenuating KLF2 degradation and inhibiting NF-κB activity. Co-culture of EVs increased the bone volume and trabecular number, but decreased bone loss in ovariectomized rats, which could be reversed after treatment with EV-miR-15b inhibitor. Conclusion Collectively, BMSC-derived EVs loaded with miR-15b promoted osteogenic differentiation by impairing WWP1-mediated KLF2 ubiquitination and inactivating the NF-κB signaling pathway. Graphical abstract ![]()
Collapse
Affiliation(s)
- Yanhong Li
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Jing Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Yanchao Ma
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Wenjia Du
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Haijun Feng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Kai Feng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Guangjie Li
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China
| | - Shuanke Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, 730030, Gansu Province, People's Republic of China.
| |
Collapse
|
45
|
Gu Z, Xie D, Ding R, Huang C, Qiu Y. GPR173 agonist phoenixin 20 promotes osteoblastic differentiation of MC3T3-E1 cells. Aging (Albany NY) 2020; 13:4976-4985. [PMID: 33196456 PMCID: PMC7950309 DOI: 10.18632/aging.103717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/29/2020] [Indexed: 11/25/2022]
Abstract
Osteogenic differentiation is critical to bone homeostasis, and its imbalance plays a key role in the progression of osteoporosis. Osteoblast cells are responsible for synthesizing new bone tissue, and understanding how to control osteoblastic differentiation is vital to the treatment of osteoporosis. Herein, we show that GPR173 signaling is involved in the regulation of osteoblastic differentiation in MC3T3-E1 cells. Our data reveals that GPR173 is abundantly expressed in MC3T3-E1 cells, and its expression is inducible upon the introduction of osteogenic media. The activation of GPR173 by its selective agonist phoenixin 20 induces the expression of several osteoblast signature genes including collagen type 1 alpha 1 (Col-I), osteocalcin (OCN), alkaline phosphatase (ALP) as well as increased matrix mineralization and ALP activity, suggesting that the activation of GPR173 promotes osteoblastic differentiation. Moreover, we show that the effect of phoenixin 20 is mediated by its induction on the key regulator runt-Related Transcription Factor 2 (Runx2). Mechanistically, we display that the action of phoenixin 20 requires the activation of MAPK kinase p38, and deactivation of p38 by its inhibitor SB203580 weakens the phoenixin 20-mediated induction of RUNX-2, ALP, and matrix mineralization. Silencing of GPR173 attenuates phoenixin 20-mediated osteoblastic differentiation, indicating its dependence on the receptor. Collectively, our study reveals a new role of GPR173 and its agonist phoenixin 20 in osteoblastic differentiation.
Collapse
Affiliation(s)
- Zhengtao Gu
- Department of Treatment Center for Traumatic Injuries, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Denghui Xie
- Division of Joint Surgery, Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Rui Ding
- Division of Spine Surgery, Section II, Department of Orthopedics, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Caiqiang Huang
- Division of Spine Surgery, Section II, Department of Orthopedics, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yiyan Qiu
- Division of Spine Surgery, Section II, Department of Orthopedics, Academy of Orthopedics of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
46
|
Hao R, Wang B, Wang H, Huo Y, Lu Y. lncRNA TUG1 promotes proliferation and differentiation of osteoblasts by regulating the miR-545-3p/CNR2 axis. ACTA ACUST UNITED AC 2020; 53:e9798. [PMID: 33053117 PMCID: PMC7552904 DOI: 10.1590/1414-431x20209798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/03/2020] [Indexed: 12/27/2022]
Abstract
Osteoblast differentiation is an effective way to promote bone formation. Long non-coding RNA taurine upregulated 1 (TUG1) has been identified as a crucial modulator of multiple biological processes. This study was designed to investigate the function of TUG1 in the proliferation and differentiation of osteoblast precursor cells hFOB1.19. In this study, we found that TUG1 promoted hFOB1.19 cell proliferation, while TUG1 knockdown hindered cell proliferation. TUG1 and cannabinoid receptor 2 (CNR2) were upregulated, while miR-545-3p was down-regulated in hFOB1.19 cells undergoing osteoblastic differentiation. TUG1 induced osteoblast differentiation by increasing alkaline phosphatase (ALP) activity and the expression of osteoblastic differentiation markers. TUG1 was a sponge of miR-545-3p and regulated osteoblastic differentiation by modulating miR-545-3p. Moreover, miR-545-3p directly targeted CNR2 and restored the effect of CNR2 on osteoblastic differentiation. In conclusion, TUG1 accelerated the proliferation and differentiation of osteoblasts by sponging miR-545-3p and increasing CNR2 expression, which might provide a new biomarker for bone diseases.
Collapse
Affiliation(s)
- Ruizheng Hao
- Department of Hand Surgery, The Second Hospital of Tangshan, Tangshan, Hebei, China
| | - Bin Wang
- Department of Hand Surgery, The Second Hospital of Tangshan, Tangshan, Hebei, China
| | - Hui Wang
- Department of Hand Surgery, The Second Hospital of Tangshan, Tangshan, Hebei, China
| | - Yongxin Huo
- Department of Hand Surgery, The Second Hospital of Tangshan, Tangshan, Hebei, China
| | - Yang Lu
- Department of Orthopedics, First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| |
Collapse
|
47
|
Valenti MT, Marchetto G, Mottes M, Dalle Carbonare L. Zebrafish: A Suitable Tool for the Study of Cell Signaling in Bone. Cells 2020; 9:E1911. [PMID: 32824602 PMCID: PMC7465296 DOI: 10.3390/cells9081911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/10/2020] [Accepted: 08/13/2020] [Indexed: 12/23/2022] Open
Abstract
In recent decades, many studies using the zebrafish model organism have been performed. Zebrafish, providing genetic mutants and reporter transgenic lines, enable a great number of studies aiming at the investigation of signaling pathways involved in the osteoarticular system and at the identification of therapeutic tools for bone diseases. In this review, we will discuss studies which demonstrate that many signaling pathways are highly conserved between mammals and teleost and that genes involved in mammalian bone differentiation have orthologs in zebrafish. We will also discuss as human diseases, such as osteogenesis imperfecta, osteoarthritis, osteoporosis and Gaucher disease can be investigated in the zebrafish model.
Collapse
Affiliation(s)
- Maria Teresa Valenti
- Department of Medicine, University of Verona, Ple Scuro 10, 37100 Verona, Italy; (G.M.); (L.D.C.)
| | - Giulia Marchetto
- Department of Medicine, University of Verona, Ple Scuro 10, 37100 Verona, Italy; (G.M.); (L.D.C.)
| | - Monica Mottes
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy;
| | - Luca Dalle Carbonare
- Department of Medicine, University of Verona, Ple Scuro 10, 37100 Verona, Italy; (G.M.); (L.D.C.)
| |
Collapse
|
48
|
Han S, Kuang M, Sun C, Wang H, Wang D, Liu Q. Circular RNA hsa_circ_0076690 acts as a prognostic biomarker in osteoporosis and regulates osteogenic differentiation of hBMSCs via sponging miR-152. Aging (Albany NY) 2020; 12:15011-15020. [PMID: 32717724 PMCID: PMC7425508 DOI: 10.18632/aging.103560] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/04/2020] [Indexed: 05/15/2023]
Abstract
OBJECTIVE Osteoporosis is the most common skeletal disease world-wide. The aim of this study is to identify potential circRNA biomarkers for osteoporosis diagnosis and treatment, as well as their roles in regulating osteogenic differentiation. RESULTS Hsa_circ_0076690 expression was significantly decreased in osteoporosis patients compared to control and showed an acceptable diagnostic value in clinical samples. Subsequently, hsa_circ_0076690 was identified to act as a sponge of miR-152. The expression of hsa_circ_0076690 was gradually increased during osteogenic differentiation while miR-152 showed a decreased expression trend. Moreover, osteogenic differentiation was promoted by hsa_circ_0076690 over-expression and remain unchanged by miR-152/hsa_circ_0076690 co-overexpression. CONCLUSIONS In conclusion, our study revealed that hsa_circ_0076690 may act as a potential diagnostic biomarker for osteoporosis patients and hsa_circ_0076690 could regulate osteogenic differentiation of hBMSCs via sponging miR-152. MATERIALS AND METHODS A total of 114 participants were enrolled in this study with ethics approvals. CircRNAs were identified by means of RNA-sequencing and qRT-PCR experiment. The clinical significance was measured by ROC curve analysis. Target relationship was validated by luciferase reporter assay. The osteogenic-associated biomarkers and ALP activity were detected by western blots.
Collapse
Affiliation(s)
- Shijie Han
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, PR China
| | - Mingjie Kuang
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, PR China
| | - Chao Sun
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, PR China
| | - Haifeng Wang
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, PR China
| | - Dachuan Wang
- Department of Orthopedics, The Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, PR China
| | - Qian Liu
- Department of Pain, Qilu Hospital of Shandong University, Jinan 250012, Sahndong, PR China
| |
Collapse
|
49
|
Shares BH, Smith CO, Sheu TJ, Sautchuk R, Schilling K, Shum LC, Paine A, Huber A, Gira E, Brown E, Awad H, Eliseev RA. Inhibition of the mitochondrial permeability transition improves bone fracture repair. Bone 2020; 137:115391. [PMID: 32360587 PMCID: PMC7354230 DOI: 10.1016/j.bone.2020.115391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/18/2022]
Abstract
Bone fracture is accompanied by trauma, mechanical stresses, and inflammation - conditions known to induce the mitochondrial permeability transition. This phenomenon occurs due to opening of the mitochondrial permeability transition pore (MPTP) promoted by cyclophilin D (CypD). MPTP opening leads to more inflammation, cell death and potentially to disruption of fracture repair. Here we performed a proof-of-concept study and tested a hypothesis that protecting mitochondria from MPTP opening via inhibition of CypD improves fracture repair. First, our in vitro experiments indicated pro-osteogenic and anti-inflammatory effects in osteoprogenitors upon CypD knock-out or pharmacological inhibition. Using a bone fracture model in mice, we observed that bone formation and biomechanical properties of repaired bones were significantly increased in CypD knock-out mice or wild type mice treated with a CypD inhibitor, NIM811, when compared to controls. These effects were evident in young male but not female mice, however in older (13 month-old) female mice bone formation was also increased during fracture repair. In contrast to global CypD knock-out, mesenchymal lineage-specific (Prx1-Cre driven) CypD deletion did not result in improved fracture repair. Our findings implicate MPTP in bone fracture and suggest systemic CypD inhibition as a modality to promote fracture repair.
Collapse
Affiliation(s)
- Brianna H Shares
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Charles O Smith
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Tzong-Jen Sheu
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Rubens Sautchuk
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14624, United States of America
| | - Laura C Shum
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Ananta Paine
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Aric Huber
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Emma Gira
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America
| | - Edward Brown
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14624, United States of America
| | - Hani Awad
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America; Department of Biomedical Engineering, University of Rochester, Rochester, NY 14624, United States of America
| | - Roman A Eliseev
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14624, United States of America; Department of Pharmacology & Physiology, University of Rochester, Rochester, NY 14624, United States of America.
| |
Collapse
|
50
|
De Santis GC, de Macedo LD, Orellana MD, Innocentini LMAR, Ferrari TC, Ricz HMA, Caruso SR, Fernandes TR, Covas DT. Mesenchymal stromal cells administration for osteonecrosis of the jaw caused by bisphosphonate: report of two cases. Acta Oncol 2020; 59:789-792. [PMID: 32079438 DOI: 10.1080/0284186x.2020.1730004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Gil Cunha De Santis
- Center for Cell-Based Therapy of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Leandro Dorigan de Macedo
- Dentistry and Stomatology Division, Hospital das Clínicas de Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Maristela Delgado Orellana
- Center for Cell-Based Therapy of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Tatiane Cristina Ferrari
- Dentistry and Stomatology Division, Hospital das Clínicas de Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Hilton Marcos Alves Ricz
- Dentistry and Stomatology Division, Hospital das Clínicas de Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sâmia Rigotto Caruso
- Center for Cell-Based Therapy of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Taísa Risque Fernandes
- Center for Cell-Based Therapy of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-Based Therapy of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Internal Medicine, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|