1
|
Thakore P, Delany AM. miRNA-based regulation in growth plate cartilage: mechanisms, targets, and therapeutic potential. Front Endocrinol (Lausanne) 2025; 16:1530374. [PMID: 40225327 PMCID: PMC11985438 DOI: 10.3389/fendo.2025.1530374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
MicroRNAs (miRNAs) are critical regulators of the skeleton. In the growth plate, these small non-coding RNAs modulate gene networks that drive key stages of chondrogenesis, including proliferation, differentiation, extracellular matrix synthesis and hypertrophy. These processes are orchestrated through the interaction of pivotal pathways including parathyroid hormone-related protein (PTHrP), Indian hedgehog (IHH), and bone morphogenetic protein (BMP) signaling. This review highlights the miRNA-mRNA target networks essential for chondrocyte differentiation. Many miRNAs are differentially expressed in resting, proliferating and hypertrophic cartilage zones. Moreover, differential enrichment of specific miRNAs in matrix vesicles is also observed, providing means for chondrocytes to influence the function and differentiation of their neighbors by via matrix vesicle protein and RNA cargo. Notably, miR-1 and miR-140 emerge as critical modulators of chondrocyte proliferation and hypertrophy by regulating multiple signaling pathways, many of them downstream from their mutual target Hdac4. Demonstration that a human gain-of-function mutation in miR-140 causes skeletal dysplasia underscores the clinical relevance of understanding miRNA-mediated regulation. Further, miRNAs such as miR-26b have emerged as markers for skeletal disorders such as idiopathic short stature, showcasing the translational relevance of miRNAs in skeletal health. This review also highlights some miRNA-based therapeutic strategies, including innovative delivery systems that could target chondrocytes via cartilage affinity peptides, and potential applications related to treatment of physeal bony bridge formation in growing children. By synthesizing current research, this review offers a nuanced understanding of miRNA functions in growth plate biology and their broader implications for skeletal health. It underscores the translational potential of miRNA-based therapies in addressing skeletal disorders and aims to inspire further investigations in this rapidly evolving field.
Collapse
|
2
|
Chen S, Bigdon S, Riether C, Ma X, Niu X, Häckel S, Li Z, Gantenbein B. The Role of the Bone Morphogenetic Protein Antagonist Noggin in Nucleus Pulposus Intervertebral Disc Cells. Int J Mol Sci 2024; 25:11803. [PMID: 39519354 PMCID: PMC11546912 DOI: 10.3390/ijms252111803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Low back pain (LBP) is a significant global health issue, contributing to disability and socioeconomic burdens worldwide. The degeneration of the human intervertebral disc (IVD) is a critical factor in the pathogenesis of LBP. Recent studies have emphasized the significance of a specific set of genes and extracellular matrix (ECM) in IVD health. In particular, Noggin has emerged as a critical gene due to its high expression levels in healthy nucleus pulposus cells (NPCs) observed in our previous research. In this study, it was hypothesized that decreased Noggin expression in NPCs is associated with IVD degeneration and contributes to LBP development. A lentivirus-mediated RNAi was applied to knock down Noggin expression in primary NPCs from six human donors. The NPCs after transduction were evaluated through cell viability analysis, XTT assay, and cell apoptosis analyses. After two weeks, a colony formation assay was used to examine the anchor-independent growth ability of transduced cells. At the transcript level, anabolic and catabolic markers were quantified using RT-qPCR. The results demonstrated that lentivirus-mediated downregulation of Noggin significantly inhibited cell proliferation, reduced cell viability, and suppressed colony formation, while inducing apoptosis in human NPCs in vitro. Notably, it disrupted cellular anabolic processes and promoted catabolic activity in human NPCs post-transduction. Our findings indicated that the degeneration of human IVD is possibly related to decreased Noggin expression in NPCs. This research provides valuable insights into the role of Noggin in IVD homeostasis and its implications in LBP treatment.
Collapse
Affiliation(s)
- Shuimu Chen
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, CH-3008 Bern, Switzerland;
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, CH-3012 Bern, Switzerland
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
| | - Sebastian Bigdon
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
| | - Carsten Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland;
- Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland; (X.M.); (X.N.)
| | - Xiaochi Ma
- Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland; (X.M.); (X.N.)
| | - Xiaoyi Niu
- Department for BioMedical Research, University of Bern, CH-3008 Bern, Switzerland; (X.M.); (X.N.)
| | - Sonja Häckel
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
- Graduate School for Health Sciences (GSH), University of Bern, CH-3012 Bern, Switzerland
| | - Zhen Li
- AO Research Institute Davos, CH-7270 Davos, Switzerland;
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopedics & Mechanobiology (TOM), Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, CH-3008 Bern, Switzerland;
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, CH-3010 Bern, Switzerland; (S.B.); (S.H.)
| |
Collapse
|
3
|
Qian F, Chen X, Wang S, Zhong Y, Liu M, Wang G, Yang X, Cheng X. MiR-322-5p is involved in regulating chondrocyte proliferation and differentiation in offspring's growth plate of maternal gestational diabetes. Sci Rep 2024; 14:20136. [PMID: 39209899 PMCID: PMC11362158 DOI: 10.1038/s41598-024-69523-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Pregestational diabetes mellitus (PGDM) has an impact on fetal bone formation, but the underlying mechanism is still obscure. Although miRNAs have been extensively investigated throughout bone formation, their effects on fetal bone development caused by PGDM still need clarification. This study intends to examine the mechanism by which hyperglycemia impairs the bone formation of offspring via miR-322-5p (miR-322). In this study, miR-322 was selected by systemically screening utilizing bioinformatics and subsequent validation experiments. Using streptozotocin (STZ)-induced diabetic mice and ATDC5 cell lines, we found that miR-322 was abundantly expressed in the proliferative and hypertrophic zones of the growth plate, and its expression pattern was disturbed in the presence of hyperglycemia, suggesting that miR-322 is involved in the chondrocyte proliferation and differentiation in absence/presence of hyperglycemia. This observation was proved by manipulating miR-322 expression in ATDC5 cells by transfecting mimic and inhibitor of miR-322. Furthermore, Adamts5, Col12a1, and Cbx6 were identified as the potential target genes of miR-322, verified by the co-transfection of miR-322 inhibitor and the siRNAs, respectively. The evaluation criteria are the chondrocyte proliferation and differentiation and their relevant key gene expressions (proliferation: Sox9 and PthIh; differentiation: Runx2 and Col10a1) after manipulating the gene expressions in ATDC5 cells. This study revealed the regulative role miR-322 on chondrocyte proliferation and differentiation of growth plate by targeting Adamts5, Col12a1, and Cbx6 in hyperglycemia during pregnancy. This translational potential represents a promising avenue for advancing our understanding of bone-related complications in diabetic pregnancy and mitigating bone deficiencies in diabetic pregnant individuals, improving maternal and fetal outcomes.
Collapse
Affiliation(s)
- Fan Qian
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Xianlong Chen
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Simiao Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Yeyin Zhong
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Min Liu
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Fujian, 350108, China
| | - Guang Wang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China
| | - Xuesong Yang
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China.
- Clinical Research Center, Clifford Hospital, Guangzhou, 511495, China.
| | - Xin Cheng
- Division of Histology and Embryology, International Joint Laboratory for Embryonic Development and Prenatal Medicine, Medical College, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
4
|
Dai T, Qiu S, Gao X, Zhao C, Ge Z, Yang Y, Tang C, Feng S. Circular RNA circWNK1 inhibits the progression of gastric cancer via regulating the miR-21-3p/SMAD7 axis. Cancer Sci 2024; 115:974-988. [PMID: 38287200 PMCID: PMC10921006 DOI: 10.1111/cas.16067] [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: 10/31/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/31/2024] Open
Abstract
Gastric cancer (GC) is a highly aggressive malignancy with limited treatment options for advanced-stage patients. Recent studies have highlighted the role of circular RNA (circRNA) as a novel regulator of cancer progression in various malignancies. However, the underlying mechanisms by which circRNA contributes to the development and progression of GC remain poorly understood. In this study, we utilized microarrays and real-time quantitative polymerase chain reaction (qRT-PCR) to identify and validate a downregulated circRNA, hsa_circ_0003251 (referred to as circWNK1), in paired GC and normal tissues. Through a series of in vitro and in vivo gain-of-function and loss-of-function assays, we demonstrated that circWNK1 exerts inhibitory effects on the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of GC cells. Additionally, we discovered that circWNK1 acts as a competitive endogenous RNA (ceRNA) for SMAD7 by sequestering miR-21-3p. Our findings were supported by comprehensive biological information analysis, as well as RNA pull-down, luciferase reporter gene, and western blot assays. Notably, the downregulation of circWNK1 in GC cells resulted in reduced SMAD7 expression, subsequently activating the TGF-β signaling pathway. Collectively, our study reveals that circWNK1 functions as a tumor suppressor in GC by regulating the miR-21-3p/SMAD7-mediated TGF-β signaling pathway. Furthermore, circWNK1 holds promise as a potential biomarker for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Ting Dai
- Department of Gastroenterology, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| | - Shengkui Qiu
- Department of General Surgery, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| | - Xuesong Gao
- Department of General Surgery, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| | - Chengjin Zhao
- Department of Gastroenterology, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| | - Zhenming Ge
- Department of Gastroenterology, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| | - Yanmei Yang
- Department of Gastroenterology, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| | - Chong Tang
- Department of General Surgery, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
- Nantong Clinical Medical CollegeKangda College of Nanjing Medical UniversityNantongChina
| | - Shichun Feng
- Department of General Surgery, Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantongChina
| |
Collapse
|
5
|
Ji X, Liu X, Li X, Du X, Fan L. MircoRNA-322-5p promotes lipopolysaccharide-induced acute kidney injury mouse models and mouse primary proximal renal tubular epithelial cell injury by regulating T-box transcription factor 21/mitogen-activated protein kinase/extracellular signal-related kinase axis. Nefrologia 2023; 43 Suppl 2:8-20. [PMID: 37179213 DOI: 10.1016/j.nefroe.2023.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/25/2023] [Indexed: 05/15/2023] Open
Abstract
INTRODUCTION AND OBJECTIVES Acute kidney injury (AKI) is a common devastating complication characterized by an abrupt loss of renal function. It is of great significance to explore promising biomarkers for AKI treatment. MATERIALS AND METHODS Here, we established LPS (lipopolysaccharide)-induced AKI mice models and LPS-induced AKI mouse renal tubular epithelial cell model. The severity of AKI was determined by the levels of BUN (blood urea nitrogen) and SCr (serum creatinine), the observation of pathological section as well as the renal tubular injury score. The apoptosis was determined by the measurement of Caspase-3 and Caspase-9 activities, and cell apoptosis assays. qRT-PCR (quantitative real-time PCR) and western blot revealed that miR-322-5p (microRNA-322-5p) was up-regulated in LPS -induced AKI models while Tbx21 (T-box transcription factor 21) was down-regulated in LPS-induced AKI models. Dual-luciferase reporter and RNA pulldown assays detected the interaction of Tbx21 with miR-322-5p. RESULTS We found that miR-322-5p was overtly over-expressed in the in vitro LPS-induced AKI model and promoted the apoptosis of AKI mouse renal tubular epithelial cells via inhibiting Tbx21, which suppressed the mitochondrial fission and cell apoptosis through MAPK/ERK (mitogen-activated protein kinase/extracellular signal-related kinase) pathway. CONCLUSIONS We demonstrated that miR-322-5p promotes LPS-induced mouse AKI by regulating Tbx21/MAPK/ERK axis, which might provide new sights for AKI research.
Collapse
Affiliation(s)
- Xiaobing Ji
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Xiaodong Liu
- Department of Nephrology, The Second People's Hospital of Lianyungang,Affiliated to Kangda College of Nanjing Medical University, Lianyungang 222023, Jiangsu, China
| | - Xiangxiang Li
- Department of Nephrology, Nanjing Yuhua Hospital, Yuhua Branch of Nanjing First Hospital, Nanjing 210039, Jiangsu, China
| | - Xin Du
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China
| | - Li Fan
- Department of Nephrology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu, China.
| |
Collapse
|
6
|
Yu L, Wen H, Liu C, Wang C, Yu H, Zhang K, Han Q, Liu Y, Han Z, Li Z, Liu N. Embryonic stem cell-derived extracellular vesicles rejuvenate senescent cells and antagonize aging in mice. Bioact Mater 2023; 29:85-97. [PMID: 37449253 PMCID: PMC10336196 DOI: 10.1016/j.bioactmat.2023.06.011] [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: 12/13/2022] [Revised: 05/11/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Aging is a degenerative process that leads to tissue dysfunction and death. Embryonic stem cells (ESCs) have great therapeutic potential for age-related diseases due to their capacity for self-renewal and plasticity. However, the use of ESCs in clinical treatment is limited by immune rejection, tumourigenicity and ethical issues. ESC-derived extracellular vesicles (EVs) may provide therapeutic effects that are comparable to those of ESCs while avoiding unwanted effects. Here, we fully evaluate the role of ESC-EVs in rejuvenation in vitro and in vivo. Using RNA sequencing (RNA-Seq) and microRNA sequencing (miRNA-Seq) screening, we found that miR-15b-5p and miR-290a-5p were highly enriched in ESC-EVs, and induced rejuvenation by silencing the Ccn2-mediated AKT/mTOR pathway. These results demonstrate that miR-15b-5p and miR-290a-5p function as potent activators of rejuvenation mediated by ESC-EVs. The rejuvenating effect of ESC-EVs was further investigated in vivo by injection into aged mice. The results showed that ESC-EVs successfully ameliorated the pathological age-related phenotypes and rescued the transcriptome profile of aged mice. Our findings demonstrate that ESC-EVs treatment can rejuvenate senescence both in vitro and in vivo and suggest the therapeutic potential of ESC-EVs as a novel cell-free alternative to ESCs for age-related diseases.
Collapse
Affiliation(s)
- Lu Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Hang Wen
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Huaxin Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Kaiyue Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qingsheng Han
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yue Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhongchao Han
- Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Na Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University, Tianjin, 300071, China
| |
Collapse
|
7
|
Yang X, Lin Y, Chen T, Hu W, Li P, Qiu X, Yang B, Liang A, Gao W. YTHDF1 Enhances Chondrogenic Differentiation by Activating the Wnt/β-Catenin Signaling Pathway. Stem Cells Dev 2023; 32:115-130. [PMID: 36647682 DOI: 10.1089/scd.2022.0216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cartilage is derived from the chondrogenic differentiation of stem cells, for which the regulatory mechanism has not been fully elucidated. N6-methyladenosine (m6A) messenger RNA (mRNA) methylation is the most common posttranscriptional modification in eukaryotic mRNAs and is mediated by m6A regulators. However, whether m6A regulators play roles in chondrogenic differentiation is unknown. Herein, we aim to determine the role of a main m6A reader protein, YTH N6-methyladenosine RNA binding protein 1 (YTHDF1), in chondrogenic differentiation regulation. Western blotting (WB) assays found that the expression of YTHDF1 increased during chondrogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). The results of quantitative polymerase chain reaction, WB, immunohistochemistry, and Alcian blue staining revealed that overexpression of YTHDF1 increased cartilage matrix synthesis and the expression of chondrogenic markers when hBMSCs, ATDC5 cells, or C3H10T1/2 cells were induced to undergo chondrogenesis. Conversely, chondrogenesis was clearly inhibited when YTHDF1 was knocked down in hBMSCs, ATDC5 cells, or C3H10T1/2 cells. Further RNA sequencing and molecular biology experiments found that YTHDF1 activated the Wnt/β-catenin signaling pathway during chondrogenic differentiation. Finally, the effects of overexpression and knockdown of YTHDF1 on chondrogenic differentiation were reversed by inhibiting or activating β-catenin activity. Therefore, we demonstrated that YTDHF1 promoted chondrogenic differentiation through activation of the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiaoming Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, P. R. China.,Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Youxi Lin
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Taiqiu Chen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Wenjun Hu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Pengfei Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Xuemei Qiu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Bo Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, P. R. China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| | - Wenjie Gao
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
8
|
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
|