1
|
Yang Y, Gao N, Ji G, Hu W, Bi R, Liang J, Liu Y. Static magnetic field contributes to osteogenic differentiation of hPDLSCs through the H19/Wnt/β-catenin axis. Gene 2025; 933:148967. [PMID: 39341520 DOI: 10.1016/j.gene.2024.148967] [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: 05/04/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Static magnetic field (SMF) as an effective physical stimulus is capable of osteogenic differentiation for multiple mesenchymal stem cells, including human periodontal ligament stem cells (hPDLSCs). However, the exact molecular mechanism is still unknown. Therefore, this study intends to excavate molecular mechanisms related to SMF in hPDLSCs using functional experiments. METHODS hPDLSCs were treated with different intensities of SMF, H19 lentivirus, and Wnt/β-catenin pathway inhibitor (XAV939). Changes in osteogenic markers (Runx2, Col Ⅰ, and BMP2), Wnt/β-catenin markers (β-catenin and GSK-3β), and calcified nodules were examined using RT-qPCR, western blotting, and alizarin red staining in hPDLSCs. RESULTS SMF upregulated the expression of H19, and SMF and overexpressing H19 facilitated the expression of osteogenic markers (Runx2, Col Ⅰ, and BMP2), activation of the Wnt/β-catenin pathway, and mineralized sediment in hPDLSCs. Knockdown of H19 alleviated SMF function, and treatment with XAV939 limited SMF- and H19-mediated osteogenic differentiation of hPDLSCs. Notably, the expression of hsa-miR-532-3p, hsa-miR-370-3p, hsa-miR-18a-5p, and hsa-miR-483-3p in hPDLSCs was regulated by SMF, and may form an endogenous competitive RNA mechanism with H19 and β-catenin. CONCLUSION SMF contributes to the osteogenic differentiation of hPDLSCs by mediating the H19/Wnt/β-catenin pathway, and hsa-miR-532-3p, hsa-miR-370-3p, hsa-miR-18a-5p, and hsa-miR-483-3p may be the key factors in it.
Collapse
Affiliation(s)
- Yanling Yang
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, No.1088 Haiyuan Middle Road, Kunming, Yunnan 650106, China; Yunnan Key Laboratory of Stomatology, Kunming Medical University, 1168 Chunrong West Road, Kunming, Yunnan 650500, China; Center of Stomatology, Affiliated Hospital of Yunnan University, No.176 Qingnian Road, Kunming, Yunnan 650021, China
| | - Na Gao
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, No.935 Jiaoling Road Kunming, Yunnan 650118, China
| | - Guang Ji
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, No.935 Jiaoling Road Kunming, Yunnan 650118, China
| | - Wenzhu Hu
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, No.935 Jiaoling Road Kunming, Yunnan 650118, China
| | - Rong Bi
- Genetic Engineering and Vaccine Research Center, Institute of Medical Biology, Chinese Academy of Medical Science, Peking Union Medical College, No.935 Jiaoling Road Kunming, Yunnan 650118, China
| | - Jiangli Liang
- Laboratory of Vaccine Development, Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, No.935 Jiaoling Road Kunming, Yunnan 650118, China
| | - Yali Liu
- Department of Orthodontics, Kunming Medical University School and Hospital of Stomatology, No.1088 Haiyuan Middle Road, Kunming, Yunnan 650106, China; Yunnan Key Laboratory of Stomatology, Kunming Medical University, 1168 Chunrong West Road, Kunming, Yunnan 650500, China.
| |
Collapse
|
2
|
Tang C, Huang L, Guo XQ, Wang GG, Chen Z. LINC01133 promotes the osteogenic differentiation of bone marrow mesenchymal stem cells by upregulating CTNNB1 by acting as a sponge for miR-214-3p. J Orthop Surg Res 2024; 19:572. [PMID: 39285416 PMCID: PMC11406849 DOI: 10.1186/s13018-024-05053-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Osteoporosis results from decreased bone mass and disturbed bone structure. Human bone marrow mesenchymal stem cells (hBMSCs) demonstrate robust osteogenic differentiation, a critical process for bone formation. This research was designed to examine the functions of LINC01133 in osteogenic differentiation. METHODS Differentially expressed lncRNAs affecting osteogenic differentiation in hBMSCs were identified from the GEO database. A total of 74 osteoporosis patients and 70 controls were enrolled. hBMSCs were stimulated to undergo osteogenic differentiation using an osteogenic differentiation medium (OM). RT-qPCR was performed to evaluate LINC01133 levels and osteogenesis-related genes such as osteocalcin, osteopontin, and RUNX2. An alkaline phosphates (ALP) activity assay was conducted to assess osteogenic differentiation. Cell apoptosis was detected using flow cytometry. Dual luciferase reporter assay and RIP assay were employed to investigate the association between miR-214-3p and LINC01133 or CTNNB1. Loss or gain of function assays were conducted to elucidate the impact of LINC01133 and miR-214-3p on osteogenic differentiation of hBMSCs. RESULTS LINC01133 and CTNNB1 expression decreased in osteoporotic patients but increased in OM-cultured hBMSCs, whereas miR-214-3p showed an opposite trend. Depletion of LINC01133 suppressed the expression of genes associated with bone formation and ALP activity triggered by OM in hBMSCs, leading to increased cell apoptosis. Nevertheless, this suppression was partially counteracted by the reduced miR-214-3p levels. Mechanistically, LINC01133 and CTNNB1 were identified as direct targets of miR-214-3p. CONCLUSIONS Our study highlights the role of LINC01133 in positively regulating CTNNB1 expression by inhibiting miR-214-3p, thereby promoting osteogenic differentiation of BMSCs. These findings may provide valuable insights into bone regeneration in osteoporosis.
Collapse
Affiliation(s)
- Chao Tang
- Department of Orthopedics, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Lina Huang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Xiu-Quan Guo
- Department of Spinal Surgery, Zhucheng People's Hospital, Weifang, Shandong, China
| | - Gang-Gang Wang
- Department of Hand and Foot Surgery, Zhucheng People's Hospital, 59 South Ring Road, Zhucheng, Weifang, 262200, Shandong, China.
| | - Zhigang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xi'an Medical University, No. 48, Fenghao West Road, Lianhu District, Xi'an, 710000, Shaanxi, China.
| |
Collapse
|
3
|
Bonilauri B, Ribeiro AL, Spangenberg L, Dallagiovanna B. Unveiling Polysomal Long Non-Coding RNA Expression on the First Day of Adipogenesis and Osteogenesis in Human Adipose-Derived Stem Cells. Int J Mol Sci 2024; 25:2013. [PMID: 38396700 PMCID: PMC10888724 DOI: 10.3390/ijms25042013] [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: 12/12/2023] [Revised: 01/06/2024] [Accepted: 01/11/2024] [Indexed: 02/25/2024] Open
Abstract
Understanding the intricate molecular mechanisms governing the fate of human adipose-derived stem cells (hASCs) is essential for elucidating the delicate balance between adipogenic and osteogenic differentiation in both healthy and pathological conditions. Long non-coding RNAs (lncRNAs) have emerged as key regulators involved in lineage commitment and differentiation of stem cells, operating at various levels of gene regulation, including transcriptional, post-transcriptional, and post-translational processes. To gain deeper insights into the role of lncRNAs' in hASCs' differentiation, we conducted a comprehensive analysis of the lncRNA transcriptome (RNA-seq) and translatome (polysomal-RNA-seq) during a 24 h period of adipogenesis and osteogenesis. Our findings revealed distinct expression patterns between the transcriptome and translatome during both differentiation processes, highlighting 90 lncRNAs that are exclusively regulated in the polysomal fraction. These findings underscore the significance of investigating lncRNAs associated with ribosomes, considering their unique expression patterns and potential mechanisms of action, such as translational regulation and potential coding capacity for microproteins. Additionally, we identified specific lncRNA gene expression programs associated with adipogenesis and osteogenesis during the early stages of cell differentiation. By shedding light on the expression and potential functions of these polysome-associated lncRNAs, we aim to deepen our understanding of their involvement in the regulation of adipogenic and osteogenic differentiation, ultimately paving the way for novel therapeutic strategies and insights into regenerative medicine.
Collapse
Affiliation(s)
- Bernardo Bonilauri
- Stem Cell Basic Biology Laboratory (LABCET), Carlos Chagas Institute—Fiocruz/PR, Curitiba 81350-010, PR, Brazil;
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Annanda Lyra Ribeiro
- Stem Cell Basic Biology Laboratory (LABCET), Carlos Chagas Institute—Fiocruz/PR, Curitiba 81350-010, PR, Brazil;
| | - Lucía Spangenberg
- Bioinformatics Unit, Institut Pasteur de Montevideo, Montevideo 11400, Uruguay;
| | - Bruno Dallagiovanna
- Stem Cell Basic Biology Laboratory (LABCET), Carlos Chagas Institute—Fiocruz/PR, Curitiba 81350-010, PR, Brazil;
| |
Collapse
|
4
|
Wei J, Zhu X, Sun AY, Yan X, Meng X, Ge S. Long non-coding RNA FGD5 antisense RNA 1 targets Baculovirus inhibitor 5 via microRNA-497-5p to alleviate calcific aortic valve disease. Clin Hemorheol Microcirc 2024; 86:285-302. [PMID: 37355887 DOI: 10.3233/ch-221692] [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] [Indexed: 06/26/2023]
Abstract
Calcific aortic valve disease (CAVD) is featured by thickening and calcification of the aortic valve. Osteoblast differentiation is a crucial step in valve calcification. Long non-coding RNAs (LncRNAs) participate in the osteogenic differentiation of mesenchymal cells. However, the character of lncRNA FGD5 antisense RNA 1 (FGD5-AS1) in CAVD is uncertain. After collection of human aortic valve tissue samples, detection of FGD5-AS1, microRNA (miR)-497-5p and Baculovirus inhibitor 5 (BIRC5) was conducted. Valve mesenchymal cells were isolated from CAVD patients and induced to differentiate to osteoblasts, and transfected with FGD5-AS1, miR-497-5p and BIRC5 plasmids. Detection of the alkaline phosphatase activity was after osteogenic induction of human aortic valve interstitial cells (hAVICs); Detection of the degree of calcium nodules and osteoblast differentiation markers (RUNX2 and OPN) was conducted. After establishment of a mouse model of CAVD, detection of the thickness of aortic valve leaflets, and the degree of calcification of the valve leaflets, and evaluation of echocardiographic parameters were implemented. Experimental data manifested in CAVD patients, lncRNAFGD5-AS1 and BIRC5 were reduced, but miR-497-5p was elevated; Enhancing lncRNA FGD5-AS1 or repressing miR-497-5p mitigated CAVD by restraining osteogenic differentiation; LncRNA FGD5-AS1 sponged miR-497-5p to target BIRC5; Repressive BIRC5 turned around the therapeutic action of elevated FGD5-AS1 or depressed miR-497-5p on hAVICs; Enhancive FGD5-AS1 in vivo was available to reduce ApoE-/- mouse CAVD induced via high cholesterol diet. All in all, lncRNAFGD5-AS1 targets BIRC5 via miR-497-5p to alleviate CAVD.
Collapse
Affiliation(s)
- Jun Wei
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Cardiovascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - XueShuang Zhu
- Department of Cardiovascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - AYu Sun
- Department of Cardiovascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - XiaoTian Yan
- Department of Cardiovascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Xing Meng
- Department of Cardiovascular Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Zhou JQ, Wan HY, Wang ZX, Jiang N. Stimulating factors for regulation of osteogenic and chondrogenic differentiation of mesenchymal stem cells. World J Stem Cells 2023; 15:369-384. [PMID: 37342227 PMCID: PMC10277964 DOI: 10.4252/wjsc.v15.i5.369] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/21/2023] [Accepted: 03/29/2023] [Indexed: 05/26/2023] Open
Abstract
Mesenchymal stem cells (MSCs), distributed in many tissues in the human body, are multipotent cells capable of differentiating in specific directions. It is usually considered that the differentiation process of MSCs depends on specialized external stimulating factors, including cell signaling pathways, cytokines, and other physical stimuli. Recent findings have revealed other underrated roles in the differentiation process of MSCs, such as material morphology and exosomes. Although relevant achievements have substantially advanced the applicability of MSCs, some of these regulatory mechanisms still need to be better understood. Moreover, limitations such as long-term survival in vivo hinder the clinical application of MSCs therapy. This review article summarizes current knowledge regarding the differentiation patterns of MSCs under specific stimulating factors.
Collapse
Affiliation(s)
- Jia-Qi Zhou
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Hao-Yang Wan
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Zi-Xuan Wang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Nan Jiang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
6
|
Qiu X, Deng Z, Wang M, Feng Y, Bi L, Li L. Piezo protein determines stem cell fate by transmitting mechanical signals. Hum Cell 2023; 36:540-553. [PMID: 36580272 DOI: 10.1007/s13577-022-00853-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/21/2022] [Indexed: 12/30/2022]
Abstract
Piezo ion channel is a mechanosensitive protein on the cell membrane, which contains Piezo1 and Piezo2. Piezo channels are activated by mechanical forces, including stretch, matrix stiffness, static pressure, and shear stress. Piezo channels transmit mechanical signals that cause different downstream responses in the differentiation process, including integrin signaling pathway, ERK1/2 MAPK signaling pathway, Notch signaling, and WNT signaling pathway. In the fate of stem cell differentiation, scientists found differences in Piezo channel expression and found that Piezo channel expression is related to developmental diseases. Here, we briefly review the structure and function of Piezo channels and the relationship between Piezo and mechanical signals, discussing the current understanding of the role of Piezo channels in stem cell fate and associated molecules and developmental diseases. Ultimately, we believe this review will help identify the association between Piezo channels and stem cell fate.
Collapse
Affiliation(s)
- Xiaolei Qiu
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Zhuoyue Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Meijing Wang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yuqi Feng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, 130021, China
| |
Collapse
|
7
|
Wang S, Chen F, Zeng C, Gu H, Wang Z, Yu W, Wu Y, Shen H. RNA Sequencing Reveals the Expression Profiles of circRNAs and Indicates Hsa_circ_0070562 as a Pro-osteogenic Factor in Bone Marrow-Derived Mesenchymal Stem Cells of Patients With Ankylosing Spondylitis. Front Genet 2022; 13:947120. [PMID: 35873481 PMCID: PMC9299369 DOI: 10.3389/fgene.2022.947120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Recent studies have reported that circular RNAs (circRNAs) play a crucial regulatory role in a variety of human diseases. However, the roles of circRNAs in pathological osteogenesis in ankylosing spondylitis (AS) remain unclear. We conducted circRNA and miRNA expression profiling of osteogenically differentiated bone marrow-derived mesenchymal stem cells (BMSCs) of patients with AS compared with those of healthy donors (HDs) by RNA sequencing (RNA-seq). Results showed that a total of 31806 circRNAs were detected in the BMSC samples, of which 418 circRNAs were significantly differentially expressed (DE) with a fold change ≥2 and p value <0.05. Among these, 204 circRNAs were upregulated, and 214 were downregulated. GO and KEGG analyses demonstrated that the DE circRNAs were mainly involved in the regulation of biological processes of the cell matrix adhesion and the TGF-beta signaling pathway, which are closely related to AS. circRNA-miRNA interaction networks related to the TGF-beta signaling pathway were established. The results of qRT-PCR showed that has_circ_0070562 was significantly up-regulated in AS-MSCs. In vitro experiments showed that silencing of has_circ_0070562 weakened osteogenesis of AS-BMSCs. In conclusion, we identified numerous circRNAs that were dysregulated in AS-BMSCs compared with HD-BMSCs. Bioinformatic analyses suggested that these dysregulated circRNAs might play important functional roles in AS-BMSCs osteogenesis. Circ_0070562 functioned as a pro-ostegenic factor and might serve as a potential biomarker and a therapeutic target for AS.
Collapse
Affiliation(s)
- Shan Wang
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Fenglei Chen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chenying Zeng
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huimin Gu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Ziming Wang
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Wenhui Yu
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yanfeng Wu
- Center for Biotherapy, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Huiyong Shen
- Department of Orthopedics, Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
8
|
Guan S, Zhang Z, Wu J. Non-coding RNA delivery for bone tissue engineering: progress, challenges and potential solutions. iScience 2022; 25:104807. [PMID: 35992068 PMCID: PMC9385673 DOI: 10.1016/j.isci.2022.104807] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
More than 20 million individuals worldwide suffer from congenital or acquired bone defects annually. The development of bone scaffold materials that simulate natural bone for bone defect repair remains challenging. Recently, ncRNA-based therapies for bone defects have attracted increasing interest because of the great potential of ncRNAs in disease treatment. Various types of ncRNAs regulate gene expression in osteogenesis-related cells via multiple mechanisms. The delivery of ncRNAs to the site of bone loss through gene vectors or scaffolds is a potential therapeutic option for bone defect repair. Therefore, this study discusses and summarizes the regulatory mechanisms of miRNAs, siRNAs, and piRNAs in osteogenic signaling and reviews the widely used current RNA delivery vectors and scaffolds for bone defect repair. Additionally, current challenges and potential solutions of delivery scaffolds for bone defect repair are proposed, with the aim of providing a theoretical basis for their future clinical applications.
Collapse
|
9
|
Liu J, Yao Y, Huang J, Sun H, Pu Y, Tian M, Zheng M, He H, Li Z. Comprehensive analysis of lncRNA-miRNA-mRNA networks during osteogenic differentiation of bone marrow mesenchymal stem cells. BMC Genomics 2022; 23:425. [PMID: 35672672 PMCID: PMC9172120 DOI: 10.1186/s12864-022-08646-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/19/2022] [Indexed: 11/15/2022] Open
Abstract
Background Long non-coding RNA (lncRNA) plays crucial role in osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs), involving in regulation of competing endogenous RNA (ceRNA) mechanisms and conduction of signaling pathways. However, its mechanisms are poorly understood. This study aimed to investigate lncRNAs, miRNAs and mRNAs expression profiles in rat BMMSCs (rBMMSCs) osteogenic differentiation, screen the potential key lncRNA-miRNA-mRNA networks, explore the putative functions and identify the key molecules, as the basis of studying potential mechanism of rBMMSCs osteogenic differentiation driven by lncRNA, providing molecular targets for the management of bone defect. Methods High-throughput RNA sequencing (RNA-seq) was used to determine lncRNAs, miRNAs, and mRNAs expression profiles at 14-day rBMMSCs osteogenesis. The pivotal lncRNA-miRNA and miRNA-mRNA networks were predicted from sequencing data and bioinformatic analysis, and the results were exported by Cytoscape 3.9.0 software. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were used for functional exploration. Real-time quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed to validate lncRNAs, miRNAs and mRNAs. Results rBMMSCs were identified, and the osteogenic and adipogenic differentiation ability were detected. A total of 8634 lncRNAs were detected by RNA-seq, and 1524 differential expressed lncRNAs, of which 812 up-regulated and 712 down-regulated in osteo-inductive groups compared with control groups. 30 up-regulated and 61 down-regulated miRNAs, 91 miRNAs were differentially expressed in total. 2453 differentially expressed mRNAs including 1272 up-expressed and 1181 down-expressed were detected. 10 up-regulated lncRNAs were chosen to predict 21 down-regulated miRNAs and 650 up-regulated mRNAs. 49 lncRNA-miRNA and 1515 miRNA–mRNA interactive networks were constructed. GO analysis showed the most important enrichment in cell component and molecular function were “cytoplasm” and “protein binding”, respectively. Biological process related to osteogenic differentiation such as “cell proliferation”, “wound healing”, “cell migration”, “osteoblast differentiation”, “extracellular matrix organization” and “response to hypoxia” were enriched. KEGG analysis showed differentially expressed genes were mainly enriched in “PI3K-Akt signaling pathway”, “Signaling pathway regulating pluripotency of stem cells”, “cGMP-PKG signaling pathway”, “Axon guidance” and “Calcium signaling pathway”. qRT-PCR verified that lncRNA Tug1, lncRNA AABR07011996.1, rno-miR-93-5p, rno-miR-322-5p, Sgk1 and Fzd4 were consistent with the sequencing results, and 4 lncRNA-miRNA-mRNA networks based on validations were constructed, and enrichment pathways were closely related to “PI3K-Akt signaling pathway”, “Signaling pathway regulating pluripotency of stem cells” and “Wnt signaling pathway”. Conclusions lncRNAs, miRNAs and mRNAs expression profiles provide clues for future studies on their roles for BMMSCs osteogenic differentiation. Furthermore, lncRNA–miRNA–mRNA networks give more information on potential new mechanisms and targets for management on bone defect. Supplementary information The online version contains supplementary material available at 10.1186/s12864-022-08646-x.
Collapse
Affiliation(s)
- Jialin Liu
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China.,Affiliated Stomatological Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China.,Stomatology Research Institute of Xinjiang Uygur Autonomous Region, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Yuan Yao
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China.,Affiliated Stomatological Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China.,Stomatology Research Institute of Xinjiang Uygur Autonomous Region, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Jinyong Huang
- Department of Trauma Orthopedics, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Hao Sun
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Yixuan Pu
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Mengting Tian
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Meijie Zheng
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China
| | - Huiyu He
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China.
| | - Zheng Li
- Department of Prosthodontics and Implant Dentistry, The First Affiliated Hospital of Xinjiang Medical University, Xin Jiang Uygur Autonomous Region, 830054, Urumqi, China.
| |
Collapse
|
10
|
Shen HX, Liu JZ, Yan XQ, Yang HN, Hu SQ, Yan XL, Xu T, El Haj AJ, Yang Y, Lü LX. Hydrostatic pressure stimulates the osteogenesis and angiogenesis of MSCs/HUVECs co-culture on porous PLGA scaffolds. Colloids Surf B Biointerfaces 2022; 213:112419. [PMID: 35227994 DOI: 10.1016/j.colsurfb.2022.112419] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 01/04/2023]
Abstract
In native bone tissue regeneration, blood vessels, providing oxygen and nutrition for tissues, can promote the regeneration of bone and accelerate the repair of a defected area. In this study, Poly(D, L-lactic-co-glycolic acid) (PLGA) inverse opal scaffolds with high pore interconnectivity were fabricated and further modified with vascular endothelial growth factor (VEGF). The rat bone marrow derived mesenchymal stem cells (rMSCs) and human umbilical vein endothelial cells (HUVECs) were co-cultured onto the scaffolds to enhance vascularization for bone tissue regeneration. Cell attachment, viability, proliferation, and morphology were detected by cell counting kit-8 (CCK-8) assay, live and dead staining and scanning electron microscopy (SEM). Hydrostatic pressure with 0-279 KPa and 1 Hz one hour per day for 7 days was applied to tissue engineered bone constructs to investigate whether the loading stimulation can promote osteogenesis and angiogenesis mutually evaluated in parallel by multiple in vitro assays and in an in vivo chicken chorioallantoic membrane (CAM) model. The results indicated that the immobilization of VEGF can improve biocompatibility of PLGA scaffolds and promote cell attachment and proliferation. The cell-scaffold constructs showed higher CD31 expression because of the angiogenic differentiation of rMSCs in hydrostatic loading culture condition in vitro. The in vivo CAM model experiment demonstrated that hydrostatic loading stimulated angiogenic differentiation of rMSCs can accelerate tubulogenesis. Furthermore, the new capillaries formed in cell-scaffold constructs were conducive to calcium deposition in vivo.
Collapse
Affiliation(s)
- Hong-Xian Shen
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Jing-Zhi Liu
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Xiao-Qing Yan
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Hong-Ning Yang
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Shu-Qun Hu
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China
| | - Xian-Liang Yan
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China; Emergency Medicine Department of the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Tie Xu
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China; Emergency Medicine Department of the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Alicia J El Haj
- Institute of Translational Medicine, University of Birmingham, Birmingham B15 2TH, UK
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK.
| | - Lan-Xin Lü
- The Laboratory of Emergency Medicine, School of the Secondary Clinical Medicine, Xuzhou Medical University, Xuzhou 221002, China; Emergency Medicine Department of the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China.
| |
Collapse
|
11
|
Shi Q, Zheng M. Role of LINC01133 in Osteogenic Differentiation of Dental Pulp Stem Cells by Targeting miR-199b-5p. ORAL HEALTH & PREVENTIVE DENTISTRY 2022; 20:173-184. [PMID: 35481341 PMCID: PMC11641068 DOI: 10.3290/j.ohpd.b2960495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
PURPOSE Recently, increasing attention has been paid to the function of long non-coding RNAs (lncRNAs) in osteogenic differentiation (OD) of dental pulp stem cells (DPSCs). LINC01133 was reported to have a close relationship with tumorigenesis for multiple cancers, but no study has yet explored the role of LINC01133 in modulating OD of DPSCs. MATERIALS AND METHODS Alizarin red S (ARS) staining and alkaline phosphatase (ALP) staining were perfomed to assess the OD potential of DPSCs. Osteogenic markers including runt-related transcription factor 2 (RUNX2), osterix (OSX) and ALP expression levels in DPSCs were monitored by qRT-PCR and Western blot before and after cell transfection. Luciferase reporter gene assay detected the relationship between LINC01133 and miR-199b-5p. RESULTS The expression of LINC01133 was low, while miR-199b-5p was increasingly expressed during OD of DPSCs. Overexpression of LINC01133 in DPSCs resulted in decreased expression of RUNX2, OSX, ALP, DSPP and DMP1, whose expression was reversed in DPSCs after transfections of miR-199b-5p overexpression. Co-transfection of pcDNA3.1-LINC01133 and miR-199b-5p mimic led to elevated expression of RUNX2, OSX, ALP, DSPP and DMP1 compared with pcDNA3.1-LINC01133 transfection alone. LINC01133 served as a sponge of miR-199b-5p. AKT3 was verified as a downstream effector of miR-199b-5p in DPSCs. CONCLUSION LINC01133 inhibits the OD of DPSCs by upregulating AKT3 via sponging miR-199b-5p, which may act as a potential diagnostic biomarker for dentin regeneration in the dental pulp.
Collapse
Affiliation(s)
- Qiaorui Shi
- Physician, Department of Special Clinic, School and Hospital of Stomatology, Fujian Medical University, Fujian Stomatological Hospital, Fuzhou, Fujian, P.R. China. Idea, experimental design, performed the experiments, analysed the data, wrote the manuscript, read and approved the final version for publication
| | - Ming Zheng
- Chief Physician, Department of Oral Prosthetics, School and Hospital of Stomatology, Fujian Medical University, Fujian Stomatological Hospital, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Oral Diseases, Laboratory of Oral Tissue Engineering, Fujian Medical University, Fuzhou, Fujian, P.R. China. Idea, experimental design, provided crucial materials, supervised the study, read and approved the final version of the manuscript for publication
| |
Collapse
|
12
|
Mechanical Static Force Negatively Regulates Vitality and Early Skeletal Development in Zebrafish Embryos. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12062912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Skeletal system development and remodelling is regulated by several different factors, including hormones, cytokines, and mechanical forces. It is known that gravity and pressure stimulate mechanosensors on bone cells which transduce mechanical signals to chemical ones. Nevertheless, few data have been provided about the role of mechanical forces on embryo osteogenesis in vivo. Since the zebrafish is an elective model for developmental studies, in particular on bone formation and tissue mineralization, we analyzed in vivo the effects of a static mechanical force generated by a water column on fertilized zebrafish eggs. The results have shown that an increase in the hydrostatic pressure (HP) of up to 5.9% was lethal for 100% of treated embryos at 48 h post fertilization (hpf). A small decrease in length (−2%) and 49% mortality were found in the +4.4% HP embryos compared with the controls. To analyze skeletal development, we evaluated the number of mineralized vertebral bodies in the trunk at five days post fertilization. The embryos grown under +2.4% HP showed a physiological intramembranous mineralization of vertebral bodies whereas the embryos which grew with +3.4% HP showed a significant decrease in mineralization rate (−54%). Morphological analysis of cartilage and bones in embryos at +3.4% HP revealed a delay of both intramembranous and chondrogenic mineralization, respectively, in axial and head bones, whereas the chondrogenesis appeared normal. These data suggested that developing osteoblasts and different mineralization programs are sensitive to mechanical pressure when applied to early embryogenesis.
Collapse
|
13
|
Tharakan S, Khondkar S, Ilyas A. Bioprinting of Stem Cells in Multimaterial Scaffolds and Their Applications in Bone Tissue Engineering. SENSORS (BASEL, SWITZERLAND) 2021; 21:7477. [PMID: 34833553 PMCID: PMC8618842 DOI: 10.3390/s21227477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022]
Abstract
Bioprinting stem cells into three-dimensional (3D) scaffolds has emerged as a new avenue for regenerative medicine, bone tissue engineering, and biosensor manufacturing in recent years. Mesenchymal stem cells, such as adipose-derived and bone-marrow-derived stem cells, are capable of multipotent differentiation in a 3D culture. The use of different printing methods results in varying effects on the bioprinted stem cells with the appearance of no general adverse effects. Specifically, extrusion, inkjet, and laser-assisted bioprinting are three methods that impact stem cell viability, proliferation, and differentiation potential. Each printing method confers advantages and disadvantages that directly influence cellular behavior. Additionally, the acquisition of 3D bioprinters has become more prominent with innovative technology and affordability. With accessible technology, custom 3D bioprinters with capabilities to print high-performance bioinks are used for biosensor fabrication. Such 3D printed biosensors are used to control conductivity and electrical transmission in physiological environments. Once printed, the scaffolds containing the aforementioned stem cells have a significant impact on cellular behavior and differentiation. Natural polymer hydrogels and natural composites can impact osteogenic differentiation with some inducing chondrogenesis. Further studies have shown enhanced osteogenesis using cell-laden scaffolds in vivo. Furthermore, selective use of biomaterials can directly influence cell fate and the quantity of osteogenesis. This review evaluates the impact of extrusion, inkjet, and laser-assisted bioprinting on adipose-derived and bone-marrow-derived stem cells along with the effect of incorporating these stem cells into natural and composite biomaterials.
Collapse
Affiliation(s)
- Shebin Tharakan
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA; (S.T.); (S.K.)
- New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, NY 11568, USA
| | - Shams Khondkar
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA; (S.T.); (S.K.)
- Department of Bioengineering, New York Institute of Technology, Old Westbury, NY 11568, USA
| | - Azhar Ilyas
- Bio-Nanotechnology and Biomaterials (BNB) Lab, New York Institute of Technology, Old Westbury, NY 11568, USA; (S.T.); (S.K.)
- Department of Electrical and Computer Engineering, New York Institute of Technology, Old Westbury, NY 11568, USA
| |
Collapse
|