1
|
Wei Z, Babkirk K, Chen S, Pei M. Epithelial-to-mesenchymal transition transcription factors: New strategies for mesenchymal tissue regeneration. Cytokine Growth Factor Rev 2025:S1359-6101(25)00032-2. [PMID: 40011185 DOI: 10.1016/j.cytogfr.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
The epithelial-mesenchymal transition transcription factors (EMT-TFs)-ZEB, SNAI, and TWIST families-have been extensively studied in embryonic development and tumor metastasis, providing valuable insight into their roles in cell behavior and transformation. These EMT-TFs have garnered increasing attention in the context of mesenchymal tissue regeneration, potentially contributing an approach for cell therapy. Given that dysregulated EMT-TF expression can impair cell survival and lineage differentiation, controlled regulation of their expression could offer significant advantages for tissue regeneration. However, there is a lack of comprehensive reviews to summarize the influence of the EMT-TFs on mesenchymal tissue regeneration and potential molecular mechanisms. This review explores the regulatory roles of ZEB, SNAI, and TWIST in the regeneration of bone, adipose, cartilage, muscle, and other mesenchymal tissues, with a focus on the underlying molecular signaling mechanisms. Gaining a deeper understanding of how EMT-TFs regulate cell proliferation, apoptosis, migration, and differentiation may offer new insights into the management of mesenchymal tissue repair and open novel avenues for enhancing tissue regeneration.
Collapse
Affiliation(s)
- Zhixin Wei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Southwest Jiaotong University, Chengdu, Sichuan 610031, China
| | - Kiya Babkirk
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506, USA
| | - Song Chen
- Department of Orthopaedics, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China; Pancreatic Injury and Repair Key Laboratory of Sichuan Province, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, China.
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
2
|
Deng LQ, Li SY, Xie T, Zeng WQ, Wang YY, Shi CJ, Jin-Fang Z. LincROR promotes tumor growth of colorectal cancer through the miR-145/WNT2B/WNT10A/Wnt/β-catenin regulatory axis. PLoS One 2024; 19:e0312417. [PMID: 39546475 PMCID: PMC11567539 DOI: 10.1371/journal.pone.0312417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/05/2024] [Indexed: 11/17/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent form of malignant tumor, and the current clinical treatments are far from satisfactory. Identifying new therapeutic targets is therefore essential for clinical practices. The long intergenic non-protein coding RNA lincROR has been shown to play a significant role in the tumorigenesis of various cancers. However, the molecular mechanism underlying lincROR-mediated CRC tumorigenesis remains unclear. In the present study, we found that knockdown of lincROR significantly inhibited cell viability in vitro, while its overexpression promoted tumor growth in vivo. Mechanistically, lincROR acted as a miRNA sponge for miR-145, thereby elevating the expression of the target genes WNT2B and WNT10A. The overexpression of WNT2B and WNT10A definitely activated the Wnt/β-catenin pathway, thus led to promoting tumorigenesis in CRC. In summary, our findings identified lincROR as a novel activator of the Wnt/β-catenin pathway by serving as a miRNA sponge for miR-145 and facilitating tumorigenesis, which suggests that lincROR may be a potential therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Li-Qiang Deng
- Shenzhen Traditional Chinese Medicine Oncology Center, Shenzhen, Guangdong, P. R. China
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, P. R. China
| | - Shi-Ying Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Tian Xie
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, P. R. China
| | - Wei-Qiang Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Yu-Yan Wang
- Shenzhen Traditional Chinese Medicine Oncology Center, Shenzhen, Guangdong, P. R. China
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, P. R. China
| | - Chuan-Jian Shi
- Shenzhen Traditional Chinese Medicine Oncology Center, Shenzhen, Guangdong, P. R. China
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, P. R. China
| | - Zhang Jin-Fang
- Shenzhen Traditional Chinese Medicine Oncology Center, Shenzhen, Guangdong, P. R. China
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, P. R. China
| |
Collapse
|
3
|
Inchingolo AM, Inchingolo AD, Nardelli P, Latini G, Trilli I, Ferrante L, Malcangi G, Palermo A, Inchingolo F, Dipalma G. Stem Cells: Present Understanding and Prospects for Regenerative Dentistry. J Funct Biomater 2024; 15:308. [PMID: 39452606 PMCID: PMC11508604 DOI: 10.3390/jfb15100308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Regenerative medicine in dentistry focuses on repairing damaged oral tissues using advanced tools like stem cells, biomaterials, and tissue engineering (TE). Mesenchymal stem cells (MSCs) from dental sources, such as dental pulp and periodontal ligament, show significant potential for tissue regeneration due to their proliferative and differentiative abilities. This systematic review, following PRISMA guidelines, evaluated fifteen studies and identified effective strategies for improving dental, periodontal, and bone tissue regeneration through scaffolds, secretomes, and bioengineering methods. Key advancements include the use of dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) to boost cell viability and manage inflammation. Additionally, pharmacological agents like matrine and surface modifications on biomaterials improve stem cell adhesion and promote osteogenic differentiation. By integrating these approaches, regenerative medicine and TE can optimize dental therapies and enhance patient outcomes. This review highlights the potential and challenges in this field, providing a critical assessment of current research and future directions.
Collapse
Affiliation(s)
- Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Paola Nardelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giulia Latini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Irma Trilli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Laura Ferrante
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK;
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (A.D.I.); (P.N.); (G.L.); (I.T.); (L.F.); (G.D.)
| |
Collapse
|
4
|
Amroodi MN, Maghsoudloo M, Amiri S, Mokhtari K, Mohseni P, Pourmarjani A, Jamali B, Khosroshahi EM, Asadi S, Tabrizian P, Entezari M, Hashemi M, Wan R. Unraveling the molecular and immunological landscape: Exploring signaling pathways in osteoporosis. Biomed Pharmacother 2024; 177:116954. [PMID: 38906027 DOI: 10.1016/j.biopha.2024.116954] [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: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Osteoporosis, characterized by compromised bone density and microarchitecture, represents a significant global health challenge, particularly in aging populations. This comprehensive review delves into the intricate signaling pathways implicated in the pathogenesis of osteoporosis, providing valuable insights into the pivotal role of signal transduction in maintaining bone homeostasis. The exploration encompasses cellular signaling pathways such as Wnt, Notch, JAK/STAT, NF-κB, and TGF-β, all of which play crucial roles in bone remodeling. The dysregulation of these pathways is a contributing factor to osteoporosis, necessitating a profound understanding of their complexities to unveil the molecular mechanisms underlying bone loss. The review highlights the pathological significance of disrupted signaling in osteoporosis, emphasizing how these deviations impact the functionality of osteoblasts and osteoclasts, ultimately resulting in heightened bone resorption and compromised bone formation. A nuanced analysis of the intricate crosstalk between these pathways is provided to underscore their relevance in the pathophysiology of osteoporosis. Furthermore, the study addresses some of the most crucial long non-coding RNAs (lncRNAs) associated with osteoporosis, adding an additional layer of academic depth to the exploration of immune system involvement in various types of osteoporosis. Finally, we propose that SKP1 can serve as a potential biomarker in osteoporosis.
Collapse
Affiliation(s)
- Morteza Nakhaei Amroodi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shayan Amiri
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Parnaz Mohseni
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Pourmarjani
- Department of Pediatrics, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behdokht Jamali
- Department of microbiology and genetics, kherad Institute of higher education, Busheher, lran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pouria Tabrizian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
5
|
Sadowska JM, Power RN, Genoud KJ, Matheson A, González-Vázquez A, Costard L, Eichholz K, Pitacco P, Hallegouet T, Chen G, Curtin CM, Murphy CM, Cavanagh B, Zhang H, Kelly DJ, Boccaccini AR, O'Brien FJ. A Multifunctional Scaffold for Bone Infection Treatment by Delivery of microRNA Therapeutics Combined With Antimicrobial Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307639. [PMID: 38009631 DOI: 10.1002/adma.202307639] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Treating bone infections and ensuring bone repair is one of the greatest global challenges of modern orthopedics, made complex by antimicrobial resistance (AMR) risks due to long-term antibiotic treatment and debilitating large bone defects following infected tissue removal. An ideal multi-faceted solution would will eradicate bacterial infection without long-term antibiotic use, simultaneously stimulating osteogenesis and angiogenesis. Here, a multifunctional collagen-based scaffold that addresses these needs by leveraging the potential of antibiotic-free antimicrobial nanoparticles (copper-doped bioactive glass, CuBG) to combat infection without contributing to AMR in conjunction with microRNA-based gene therapy (utilizing an inhibitor of microRNA-138) to stimulate both osteogenesis and angiogenesis, is developed. CuBG scaffolds reduce the attachment of gram-positive bacteria by over 80%, showcasing antimicrobial functionality. The antagomiR-138 nanoparticles induce osteogenesis of human mesenchymal stem cells in vitro and heal a large load-bearing defect in a rat femur when delivered on the scaffold. Combining both promising technologies results in a multifunctional antagomiR-138-activated CuBG scaffold inducing hMSC-mediated osteogenesis and stimulating vasculogenesis in an in vivo chick chorioallantoic membrane model. Overall, this multifunctional scaffold catalyzes killing mechanisms in bacteria while inducing bone repair through osteogenic and angiogenic coupling, making this platform a promising multi-functional strategy for treating and repairing complex bone infections.
Collapse
Affiliation(s)
- Joanna M Sadowska
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Rachael N Power
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Katelyn J Genoud
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
| | - Austyn Matheson
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
| | - Arlyng González-Vázquez
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Lara Costard
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Kian Eichholz
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Pierluca Pitacco
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Tanguy Hallegouet
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- University of Strasbourg, Strasbourg, 67412, France
| | - Gang Chen
- Microsurgical Research and Training Facility (MRTF), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Ciara M Murphy
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
| | - Huijun Zhang
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91056, Erlangen, Germany
| | - Daniel J Kelly
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg, 91056, Erlangen, Germany
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences, Dublin, D02 YN77, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland (RCSI) University of Medicine and Health Sciences and Trinity College Dublin (TCD), Dublin, D02 W085, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin, D02 R590, Ireland
| |
Collapse
|
6
|
Li B, Liu S, He Z, Luo E, Liu H. The role of zinc finger proteins in the fate determination of mesenchymal stem cells during osteogenic and adipogenic differentiation. Int J Biochem Cell Biol 2024; 167:106507. [PMID: 38142772 DOI: 10.1016/j.biocel.2023.106507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/26/2023]
Abstract
Zinc finger proteins (ZFPs) constitute a crucial group of transcription factors widely present in various organisms. They act as transcription factors, nucleases, and RNA-binding proteins, playing significant roles in cell differentiation, growth, and development. With extensive research on ZFPs, their roles in the determination of mesenchymal stem cells (MSCs) fate during osteogenic and adipogenic differentiation processes have become increasingly clear. ZFP521, for instance, is identified as an inhibitor of the Wnt signaling pathway and RUNX2's transcriptional activity, effectively suppressing osteogenic differentiation. Moreover, ZFP217 contributes to the inhibition of adipogenic differentiation by reducing the M6A level of the cell cycle regulator cyclin D1 (CCND1). In addition, other ZFPs can also influence the fate of mesenchymal stem cells (MSCs) during osteogenic and adipogenic differentiation through various signaling pathways, transcription factors, and epigenetic controls, participating in the subsequent differentiation and maturation of precursor cells. Given the prevalent occurrence of osteoporosis, obesity, and related metabolic disorders, a comprehensive understanding of the regulatory mechanisms balancing bone and fat metabolism is essential, with a particular focus on the fate determination of MSCs in osteogenic and adipogenic differentiation. In this review, we provide a detailed summary of how zinc finger proteins influence the osteogenic and adipogenic differentiation of MSCs through different signaling pathways, transcription factors, and epigenetic mechanisms. Additionally, we outline the regulatory mechanisms of ZFPs in controlling osteogenic and adipogenic differentiation based on various stages of MSC differentiation.
Collapse
Affiliation(s)
- Bolun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Shibo Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ze He
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
7
|
Baniasadi M, Talebi S, Mokhtari K, Zabolian AH, Khosroshahi EM, Entezari M, Dehkhoda F, Nabavi N, Hashemi M. Role of non-coding RNAs in osteoporosis. Pathol Res Pract 2024; 253:155036. [PMID: 38134836 DOI: 10.1016/j.prp.2023.155036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/10/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023]
Abstract
Osteoporosis, a prevalent bone disorder influenced by genetic and environmental elements, significantly increases the likelihood of fractures and bone weakness, greatly affecting the lives of those afflicted. Yet, the exact epigenetic processes behind the onset of osteoporosis are still unclear. Growing research indicates that epigenetic changes could act as vital mediators that connect genetic tendencies and environmental influences, thereby increasing the risk of osteoporosis and bone fractures. Within these epigenetic factors, certain types of RNA, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been recognized as key regulatory elements. These RNA types wield significant influence on gene expression through epigenetic regulation, directing various biological functions essential to bone metabolism. This extensive review compiles current research uncovering the complex ways in which miRNAs, lncRNAs, and circRNAs are involved in the development of osteoporosis, especially in osteoblasts and osteoclasts. Gaining a more profound understanding of the roles these three RNA classes play in osteoporosis could reveal new diagnostic methods and treatment approaches for this incapacitating condition. In conclusion, this review delves into the complex domain of epigenetic regulation via non-coding RNA in osteoporosis. It sheds light on the complex interactions and mechanisms involving miRNAs, lncRNAs, and circRNAs within osteoblasts and osteoclasts, offering an in-depth understanding of the less explored aspects of osteoporosis pathogenesis. These insights not only reveal the complexity of the disease but also offer significant potential for developing new diagnostic methods and targeted treatments. Therefore, this review marks a crucial step in deciphering the elusive complexities of osteoporosis, leading towards improved patient care and enhanced quality of life.
Collapse
Affiliation(s)
- Mojtaba Baniasadi
- Department of Orthopedics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sina Talebi
- Department of Orthopedics, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran; Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan,Iran
| | - Amir Hossein Zabolian
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Farshid Dehkhoda
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Noushin Nabavi
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
8
|
Brito VGB, Bell-Hensley A, McAlinden A. MicroRNA-138: an emerging regulator of skeletal development, homeostasis, and disease. Am J Physiol Cell Physiol 2023; 325:C1387-C1400. [PMID: 37842749 PMCID: PMC10861148 DOI: 10.1152/ajpcell.00382.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/17/2023]
Abstract
Noncoding microRNAs are powerful epigenetic regulators of cellular processes by their ability to target and suppress expression of numerous protein-coding mRNAs. This multitargeting function is a unique and complex feature of microRNAs. It is now well-described that microRNAs play important roles in regulating the development and homeostasis of many cell/tissue types, including those that make up the skeletal system. In this review, we focus on microRNA-138 (miR-138) and its effects on regulating bone and cartilage cell differentiation and function. In addition to its reported role as a tumor suppressor, miR-138 appears to function as an inhibitor of osteoblast differentiation. This review provides additional information on studies that have attempted to alter miR-138 expression in vivo as a means to dampen ectopic calcification or alter bone mass. However, a review of the published literature on miR-138 in cartilage reveals a number of contradictory and inconclusive findings with respect to regulating chondrogenesis and chondrocyte catabolism. This highlights the need for more research in understanding the role of miR-138 in cartilage biology and disease. Interestingly, a number of studies in other systems have reported miR-138-mediated effects in dampening inflammation and pain responses. Future studies will reveal if a multifunctional role of miR-138 involving suppression of ectopic bone, inflammation, and pain will be beneficial in skeletal conditions such as osteoarthritis and heterotopic ossification.
Collapse
Affiliation(s)
- Victor Gustavo Balera Brito
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Austin Bell-Hensley
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospital for Children, St. Louis, Missouri, United States
| |
Collapse
|
9
|
Huang J, Shen HL, Feng ML, Li Z, An S, Cao GL. Induction of lncRNA MALAT1 by hypoxia promotes bone formation by regulating the miR-22-3p/CEBPD axis. Histol Histopathol 2023; 38:1043-1053. [PMID: 36541404 DOI: 10.14670/hh-18-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Adaptation to hypoxia promotes fracture healing. However, the underlying molecular mechanism remains unknown. Increasing evidence has indicated that long non-coding RNAs (lncRNAs) play crucial roles in several diseases, including fracture healing. In the present study, lncRNA microarray analysis was performed to assess the expression levels of different lncRNAs in MC3T3-E1 cells cultured under hypoxic conditions. A total of 42 lncRNAs exhibited significant differences in their expression, including metastasis associated lung adenocarcinoma transcript 1 (MALAT1), maternally expressed 3, AK046686, AK033442, small nucleolar RNA host gene 2 and distal-less homeobox 1 splice variant 2. Furthermore, overexpression of MALAT1 promoted osteoblast differentiation, alkaline phosphatase (ALP) activity and matrix mineralization of MC3T3-E1 cells, whereas its knockdown diminished hypoxia-induced cell differentiation, ALP activity and matrix mineralization in these cells. Moreover, functional analysis indicated that MALAT1 regulated the mRNA and protein expression levels of CCAAT/enhancer binding protein δ by competitively binding to microRNA-22-3p. Adenoviral-mediated MALAT1 knockdown inhibited fracture healing in a mouse model. Taken together, the results indicated that MALAT1 may serve a role in hypoxia-mediated osteogenesis and bone formation.
Collapse
Affiliation(s)
- Jiang Huang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hui-Liang Shen
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ming-Li Feng
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zheng Li
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuai An
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Guang-Lei Cao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
An F, Wang X, Wang C, Liu Y, Sun B, Zhang J, Gao P, Yan C. Research progress on the role of lncRNA-miRNA networks in regulating adipogenic and osteogenic differentiation of bone marrow mesenchymal stem cells in osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1210627. [PMID: 37645421 PMCID: PMC10461560 DOI: 10.3389/fendo.2023.1210627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 07/26/2023] [Indexed: 08/31/2023] Open
Abstract
Osteoporosis (OP) is characterized by a decrease in osteoblasts and an increase in adipocytes in the bone marrow compartment, alongside abnormal bone/fat differentiation, which ultimately results in imbalanced bone homeostasis. Bone marrow mesenchymal stem cells (BMSCs) can differentiate into osteoblasts and adipocytes to maintain bone homeostasis. Several studies have shown that lncRNAs are competitive endogenous RNAs that form a lncRNA-miRNA network by targeting miRNA for the regulation of bone/fat differentiation in BMSCs; this mechanism is closely related to the corresponding treatment of OP and is important in the development of novel OP-targeted therapies. However, by reviewing the current literature, it became clear that there are limited summaries discussing the effects of the lncRNA-miRNA network on osteogenic/adipogenic differentiation in BMSCs. Therefore, this article provides a review of the current literature to explore the impact of the lncRNA-miRNA network on the osteogenic/adipogenic differentiation of BMSCs, with the aim of providing a new theoretical basis for the treatment of OP.
Collapse
Affiliation(s)
- Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiaxia Wang
- School of Tradional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunmei Wang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Ying Liu
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Bai Sun
- School of Tradional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Jie Zhang
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Peng Gao
- School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Chunlu Yan
- School of Tradional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Shi CJ, Lv MY, Deng LQ, Zeng WQ, Fu WM, Zhang JF. Linc-ROR drive adriamycin resistance by targeting AP-2α/Wnt/β-catenin axis in hepatocellular carcinoma. Cell Biol Toxicol 2023; 39:1735-1752. [PMID: 36576707 DOI: 10.1007/s10565-022-09777-3] [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: 06/24/2022] [Accepted: 11/11/2022] [Indexed: 12/29/2022]
Abstract
Adriamycin is widely used as a chemotherapeutic strategy for advanced hepatocellular carcinoma (HCC). However, the clinical response was disappointing because of the acquired drug resistance with long-term usage. Revealing the underlying mechanism could provide promising therapeutics for the drug-resistant patients. The recently identified linc-ROR (long intergenic non-protein-coding RNA, regulator of reprogramming) has been found to be an oncogene in various cancers, and it also demonstrated to mediate drug resistance and metastasis. We thereby wonder whether this lincRNA could mediate adriamycin chemoresistance in HCC. In this study, linc-ROR was found to be upregulated in adriamycin-resistant HCC cells. And its overexpression accelerated epithelial-mesenchymal transition (EMT) program and adriamycin resistance. Conversely, its silence suppressed EMT and made HCC cells sensitize to adriamycin in vitro and in vivo. Further investigation revealed that linc-ROR physically interacted with AP-2α, mediated its stability by a post-translational modification manner, and sequentially activated Wnt/β-catenin pathway. Furthermore, linc-ROR expression was positively associated with β-catenin expression in human clinical specimens. Taken together, linc-ROR promoted tumorigenesis and adriamycin resistance in HCC via a linc-ROR/AP-2α/Wnt/β-catenin axis, which could be developed as a potential therapeutic target for the adriamycin-resistant patients.
Collapse
Affiliation(s)
- Chuan-Jian Shi
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Min-Yi Lv
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Pharmacy, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, 518028, People's Republic of China
| | - Li-Qiang Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wei-Qiang Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Wei-Ming Fu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Jin-Fang Zhang
- Cancer Center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, 518000, People's Republic of China.
| |
Collapse
|
12
|
Sherazi SAM, Abbasi A, Jamil A, Uzair M, Ikram A, Qamar S, Olamide AA, Arshad M, Fried PJ, Ljubisavljevic M, Wang R, Bashir S. Molecular hallmarks of long non-coding RNAs in aging and its significant effect on aging-associated diseases. Neural Regen Res 2023; 18:959-968. [PMID: 36254975 PMCID: PMC9827784 DOI: 10.4103/1673-5374.355751] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 01/11/2023] Open
Abstract
Aging is linked to the deterioration of many physical and cognitive abilities and is the leading risk factor for Alzheimer's disease. The growing aging population is a significant healthcare problem globally that researchers must investigate to better understand the underlying aging processes. Advances in microarrays and sequencing techniques have resulted in deeper analyses of diverse essential genomes (e.g., mouse, human, and rat) and their corresponding cell types, their organ-specific transcriptomes, and the tissue involved in aging. Traditional gene controllers such as DNA- and RNA-binding proteins significantly influence such programs, causing the need to sort out long non-coding RNAs, a new class of powerful gene regulatory elements. However, their functional significance in the aging process and senescence has yet to be investigated and identified. Several recent researchers have associated the initiation and development of senescence and aging in mammals with several well-reported and novel long non-coding RNAs. In this review article, we identified and analyzed the evolving functions of long non-coding RNAs in cellular processes, including cellular senescence, aging, and age-related pathogenesis, which are the major hallmarks of long non-coding RNAs in aging.
Collapse
Affiliation(s)
- Syed Aoun Mehmood Sherazi
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University, Islamabad, Pakistan
| | - Asim Abbasi
- Department of Biological Sciences, University of Arkansas, Fayetteville, AR, USA
| | - Abdullah Jamil
- Department of Pharmacology, Government College University, Faisalabad, Pakistan
| | - Mohammad Uzair
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University, Islamabad, Pakistan
| | - Ayesha Ikram
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Shanzay Qamar
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | | | - Muhammad Arshad
- Department of Biological Sciences, Faculty of Basic & Applied Sciences, International Islamic University, Islamabad, Pakistan
| | - Peter J. Fried
- Department of Neurology, Berenson-Allen Center for Noninvasive Brain Stimulation and Division of Cognitive Neurology, Beth Israel Deaconess Medical Center (KS 158), Harvard Medical School, Boston, MA, USA
| | - Milos Ljubisavljevic
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ran Wang
- Department of Psychiatry, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
- Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Shahid Bashir
- Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| |
Collapse
|
13
|
Tani S, Okada H, Onodera S, Chijimatsu R, Seki M, Suzuki Y, Xin X, Rowe DW, Saito T, Tanaka S, Chung UI, Ohba S, Hojo H. Stem cell-based modeling and single-cell multiomics reveal gene-regulatory mechanisms underlying human skeletal development. Cell Rep 2023; 42:112276. [PMID: 36965484 DOI: 10.1016/j.celrep.2023.112276] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 01/19/2023] [Accepted: 03/02/2023] [Indexed: 03/27/2023] Open
Abstract
Although the skeleton is essential for locomotion, endocrine functions, and hematopoiesis, the molecular mechanisms of human skeletal development remain to be elucidated. Here, we introduce an integrative method to model human skeletal development by combining in vitro sclerotome induction from human pluripotent stem cells and in vivo endochondral bone formation by implanting the sclerotome beneath the renal capsules of immunodeficient mice. Histological and scRNA-seq analyses reveal that the induced bones recapitulate endochondral ossification and are composed of human skeletal cells and mouse circulatory cells. The skeletal cell types and their trajectories are similar to those of human embryos. Single-cell multiome analysis reveals dynamic changes in chromatin accessibility associated with multiple transcription factors constituting cell-type-specific gene-regulatory networks (GRNs). We further identify ZEB2, which may regulate the GRNs in human osteogenesis. Collectively, these results identify components of GRNs in human skeletal development and provide a valuable model for its investigation.
Collapse
Affiliation(s)
- Shoichiro Tani
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| | - Hiroyuki Okada
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Ryota Chijimatsu
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Masahide Seki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8562, Japan
| | - Xiaonan Xin
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - David W Rowe
- Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Taku Saito
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Sakae Tanaka
- Sensory and Motor System Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Ung-Il Chung
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinsuke Ohba
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Cell Biology, Institute of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan; Department of Oral Anatomy and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan.
| | - Hironori Hojo
- Laboratory of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan.
| |
Collapse
|
14
|
Mishra A, Kumar R, Mishra SN, Vijayaraghavalu S, Tiwari NK, Shukla GC, Gurusamy N, Kumar M. Differential Expression of Non-Coding RNAs in Stem Cell Development and Therapeutics of Bone Disorders. Cells 2023; 12:cells12081159. [PMID: 37190068 PMCID: PMC10137108 DOI: 10.3390/cells12081159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Stem cells' self-renewal and multi-lineage differentiation are regulated by a complex network consisting of signaling factors, chromatin regulators, transcription factors, and non-coding RNAs (ncRNAs). Diverse role of ncRNAs in stem cell development and maintenance of bone homeostasis have been discovered recently. The ncRNAs, such as long non-coding RNAs, micro RNAs, circular RNAs, small interfering RNA, Piwi-interacting RNAs, etc., are not translated into proteins but act as essential epigenetic regulators in stem cells' self-renewal and differentiation. Different signaling pathways are monitored efficiently by the differential expression of ncRNAs, which function as regulatory elements in determining the fate of stem cells. In addition, several species of ncRNAs could serve as potential molecular biomarkers in early diagnosis of bone diseases, including osteoporosis, osteoarthritis, and bone cancers, ultimately leading to the development of new therapeutic strategies. This review aims to explore the specific roles of ncRNAs and their effective molecular mechanisms in the growth and development of stem cells, and in the regulation of osteoblast and osteoclast activities. Furthermore, we focus on and explore the association of altered ncRNA expression with stem cells and bone turnover.
Collapse
Affiliation(s)
- Anurag Mishra
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Rishabh Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| | - Satya Narayan Mishra
- Maa Gayatri College of Pharmacy, Dr. APJ Abdul Kalam Technical University, Prayagraj 211009, India
| | | | - Neeraj Kumar Tiwari
- Department of IT-Satellite Centre, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India
| | - Girish C Shukla
- Department of Biological, Geological, and Environmental Sciences, 2121 Euclid Ave., Cleveland, OH 44115, USA
- Center for Gene Regulation in Health and Disease, 2121 Euclid Ave., Cleveland, OH 44115, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Munish Kumar
- Department of Biochemistry, Faculty of Science, University of Allahabad, Prayagraj 211002, India
| |
Collapse
|
15
|
Feng L, Yang Z, Hou N, Wang M, Lu X, Li Y, Wang H, Wang Y, Bai S, Zhang X, Lin Y, Yan X, Lin S, Tortorella MD, Li G. Long Non-Coding RNA Malat1 Increases the Rescuing Effect of Quercetin on TNFα-Impaired Bone Marrow Stem Cell Osteogenesis and Ovariectomy-Induced Osteoporosis. Int J Mol Sci 2023; 24:5965. [PMID: 36983039 PMCID: PMC10059267 DOI: 10.3390/ijms24065965] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Osteoporosis, a common systematic bone homeostasis disorder related disease, still urgently needs innovative treatment methods. Several natural small molecules were found to be effective therapeutics in osteoporosis. In the present study, quercetin was screened out from a library of natural small molecular compounds by a dual luciferase reporter system. Quercetin was found to upregulate Wnt/β-catenin while inhibiting NF-κB signaling activities, and thereby rescuing osteoporosis-induced tumor necrosis factor alpha (TNFα) impaired BMSCs osteogenesis. Furthermore, a putative functional lncRNA, Malat1, was shown to be a key mediator in quercetin regulated signaling activities and TNFα-impaired BMSCs osteogenesis, as mentioned above. In an ovariectomy (OVX)-induced osteoporosis mouse model, quercetin administration could significantly rescue OVX-induced bone loss and structure deterioration. Serum levels of Malat1 were also obviously rescued in the OVX model after quercetin treatment. In conclusion, our study demonstrated that quercetin could rescue TNFα-impaired BMSCs osteogenesis in vitro and osteoporosis-induced bone loss in vivo, in a Malat1-dependent manner, suggesting that quercetin may serve as a therapeutic candidate for osteoporosis treatment.
Collapse
Affiliation(s)
- Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Zhengmeng Yang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Nan Hou
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Ming Wang
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Xuan Lu
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Yucong Li
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Haixing Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Shanshan Bai
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Xiaoting Zhang
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Yuejun Lin
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Xu Yan
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Sien Lin
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Micky D. Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen 518000, China
| |
Collapse
|
16
|
Shi CJ, Xue ZH, Zeng WQ, Deng LQ, Pang FX, Zhang FW, Fu WM, Zhang JF. LncRNA-NEF suppressed oxaliplatin resistance and epithelial-mesenchymal transition in colorectal cancer through epigenetically inactivating MEK/ERK signaling. Cancer Gene Ther 2023:10.1038/s41417-023-00595-1. [PMID: 36782047 DOI: 10.1038/s41417-023-00595-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 01/06/2023] [Accepted: 02/01/2023] [Indexed: 02/15/2023]
Abstract
A major cause of oxaliplatin chemoresistance in colorectal cancer (CRC) is acquired epithelial-mesenchymal transition (EMT) in cancer cells, making the cancer cells easy to metastasis and recurrence. LncRNA Neighboring Enhancer of FOXA2 (lncRNA-NEF) has been characterized as a tumor suppressor to mediate cancer metastasis in multiple cancer types. However, whether it mediated the drug resistance remains unknown. In the present study, an oxaliplatin-resistant CRC cell line (SW620R) was established and lncRNA-NEF was obviously down-regulated in this resistant cell line. The further loss and gain-of-function studies demonstrated that this lncRNA suppressed oxaliplatin resistance as well as EMT programme in vitro and inhibited metastasis in vivo. Mechanistically, lncRNA-NEF epigenetically promoted the expression of DOK1 (Downstream of Tyrosine kinase 1), a negative regulator of MEK/ERK signaling, by disrupting DNA methyltransferases (DNMTs)-mediated DNA methylation. DOK1, in turn, induced the inactivation of MEK/ERK signaling, forming the lncRNA-NEF/DOK1/MEK/ERK regulatory axis to mediate oxaliplatin resistance in CRC. Collectively, our work reveals the critical function of lncRNA-NEF in mediating the oxaliplatin chemotherapy resistance in CRC, and provides a promising therapeutic strategy for CRC patients with oxaliplatin resistance.
Collapse
Affiliation(s)
- Chuan-Jian Shi
- Cancer center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, PR China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Zhi-He Xue
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Wei-Qiang Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Li-Qiang Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Feng-Xiang Pang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Feng-Wei Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Wei-Ming Fu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China.
| | - Jin-Fang Zhang
- Cancer center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, PR China.
| |
Collapse
|
17
|
Pang F, Ding S, Li N, Li Z, Tian N, Shi C, Zhang F, Mai Y, Zhang J, Wang J. Gallic acid mediates tumor-suppressive effects on osteosarcoma through the H19-Wnt/β-catenin regulatory axis. J Orthop Translat 2023; 39:34-42. [PMID: 36636358 PMCID: PMC9826808 DOI: 10.1016/j.jot.2022.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/06/2023] Open
Abstract
Background Osteosarcoma (OS) is the most common primary malignancy in bone tissues, and effective therapeutics remain absent in clinical practice. Traditional Chinese medicines (TCM) have been used for thousands of years, which provide great insights into OS management. Gallic acid (GA) is a natural phenolic acid enriched in various foods and herbs. Several pharmacological activities of GA such as anti-oxidation and anti-inflammation have been well-established. However, its biological function in OS remains not fully understood. Methods The potential anti-cancer properties of GA were evaluated in 143 B, U2OS and MG63 cells. Its effects on cell growth, cell cycle, apoptosis and migration were examined in these OS cells. The lncRNA H19 and Wnt/β-catenin signaling were detected by qPCR, luciferase activity and Western blotting assays. The in vivo effect of GA on tumor growth was investigated using an orthotopic mouse model. Results In the present study, GA was found to suppress the tumor growth in vitro via inducing cell cycle arrest and apoptosis in OS cells, and inhibit the invasion and metastasis as well. Using the orthotopic animal model, GA was also found to suppress tumorigenesis in vivo. Long noncoding RNA (lncRNA) H19 was demonstrated to be down-regulated by GA, and thus disrupted the canonical Wnt/β-catenin signaling in OS cells. Furthermore, the ectopic expression of H19 rescued the GA-induced suppressive effects on tumor growth and metastasis, and partially reversed the inactivation of Wnt/β-catenin signaling. Conclusions Taken together, our results indicated that GA inhibited tumor growth through an H19-mediated Wnt/β-catenin signaling regulatory axis in OS cells. The translational potential of this article The information gained from this study provides a novel underlying mechanism of GA mediated anti-OS activity, suggesting that GA may be a promising drug candidate for OS patients.
Collapse
Key Words
- CD44, cluster of differentiation 44
- GA, gallic acid
- Gallic acid
- H19
- IHC, Immunohistochemistry
- LncRNAs, long noncoding RNAs
- Metastasis
- Myc, Cellular-myelocytomatosis viral oncogene
- OS, osteosarcoma
- Oct3/4, POU class 5 homeobox 1
- Osteosarcoma
- PI, propidium iodide
- RIPA, Radio Immunoprecipitation Assay
- TCM, traditional Chinese medicine
- Wnt/β-catenin signaling
- pBabe, the empty lasmids
- pH19, H19 overexpression plasmids
- qRT-PCR, Quantitative reverse-transcription polymerase chain reaction
Collapse
Affiliation(s)
- Fengxiang Pang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Shouchang Ding
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Nan Li
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Zhipeng Li
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Nannan Tian
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Chuanjian Shi
- School of Pharmaceutical Sciences, Southern Medical University,Guangzhou, Guangdong, 511458, China
| | - Fengwei Zhang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Yongxin Mai
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China,The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510405, China
| | - Jinfang Zhang
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, China,Corresponding author. Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine.
| | - Junyan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China,Corresponding author. School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
18
|
Li Y, Yao X, Lin Y, Xing Y, Liu C, Xu J, Wu D. Identification and validation of autophagy-related genes during osteogenic differentiation of bone marrow mesenchymal stem cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1364-1372. [PMID: 36474568 PMCID: PMC9699953 DOI: 10.22038/ijbms.2022.65528.14420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/10/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVES Osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is an essential stage in bone formation. Autophagy plays a pivotal role in the self-renewal potential and pluripotency of stem cells. This study aimed to explore the function of autophagy-related genes during osteogenic differentiation of BMSCs. MATERIALS AND METHODS The differentially expressed autophagy-related genes (ARGs) were obtained from the GEO and HADb databases. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed using R software. The PPI and hub gene mining networks were constructed using the STRING database and Cytoscape. Finally, the RT-qPCR was conducted to validate the expression level of ARGs in BMSCs. RESULTS Thirty-seven differentially expressed ARGs were finally obtained, including 12 upregulated and 25 downregulated genes. GO and KEGG enrichment analysis showed that most of these genes were enriched in apoptosis and autophagy. The PPI network revealed strong interactions between differentially expressed ARGs. The expression level of differentially expressed ARGs tested by RT-qPCR showed 6 upregulated ARGs, including FOXO1, MAP1LC3C, CTSB, FOXO3, CALCOCO2, FKBP1A, and 4 downregulated ARGs, including MAPK8IP1, NRG1, VEGFA, and ITGA6 were consistent with the expression of high-throughput sequencing data. CONCLUSION We identified 37 ARGs during osteogenic differentiation using bioinformatics analysis. FOXO1, MAP1LC3C, CTSB, FOXO3, CALCOCO2, FKBP1A, MAPK8IP1, NRG1, VEGFA, and ITGA6 may regulate osteogenic differentiation of hBMSCs by involving autophagy pathway. This study provides new insight into the osteogenic differentiation of hBMSCs and may be available in developing therapeutic strategies for maxillofacial bone defects.
Collapse
Affiliation(s)
- Yan Li
- Provincial Engineering Research Center of Oral Biomaterial, Fujian Medical University, Fuzhou, Fujian, 350001, China,Department of Oral Implantology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, 350001, China,Research Center of Dental and Craniofacial Implants, Fujian Medical University, Fuzhou, Fujian, 350001, China,These authors contributed eqully to this work
| | - Xiu Yao
- Provincial Engineering Research Center of Oral Biomaterial, Fujian Medical University, Fuzhou, Fujian, 350001, China,Department of Implantology, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, 200433, China,These authors contributed eqully to this work
| | - Yanjun Lin
- Provincial Engineering Research Center of Oral Biomaterial, Fujian Medical University, Fuzhou, Fujian, 350001, China,Department of Oral Implantology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Yifeng Xing
- Provincial Engineering Research Center of Oral Biomaterial, Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Chaowei Liu
- Provincial Engineering Research Center of Oral Biomaterial, Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Jianghan Xu
- Provincial Engineering Research Center of Oral Biomaterial, Fujian Medical University, Fuzhou, Fujian, 350001, China
| | - Dong Wu
- Department of Oral Implantology, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, 350001, China,Research Center of Dental and Craniofacial Implants, Fujian Medical University, Fuzhou, Fujian, 350001, China,Corresponding author: Dong Wu. Research Center of Dental and Craniofacial Implants, Fujian Medical University, No. 246, Yangqiao Road, Gulou District, Fuzhou, Fujian 350001, China.
| |
Collapse
|
19
|
Zhao H, Li L, Zhao N, Lu A, Lu C, He X. The effect of long non-coding RNAs in joint destruction of rheumatoid arthritis. Front Cell Dev Biol 2022; 10:1011371. [PMID: 36263019 PMCID: PMC9574091 DOI: 10.3389/fcell.2022.1011371] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 11/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease accompanied with joint destruction. Serious joint destruction will eventually lead to disability and the decline of life quality in RA patients. At present, the therapeutic effect of drugs to alleviate joint destruction in RA is limited. Recently, accumulating evidences have shown that long non-coding RNAs (lncRNAs) play an important role in the pathogenesis of joint diseases. Therefore, this paper reviews the expression change and the action mechanism of lncRNAs in joint destruction of RA in recent years. A more comprehensive understanding of the role of lncRNAs in joint destruction will help the treatment of RA.
Collapse
Affiliation(s)
- Hanxiao Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ning Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Shanghai GuangHua Hospital of Integrated Traditional Chinese and Western Medicine, Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, China
- *Correspondence: Aiping Lu, ; Cheng Lu, ; Xiaojuan He,
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Aiping Lu, ; Cheng Lu, ; Xiaojuan He,
| | - Xiaojuan He
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Aiping Lu, ; Cheng Lu, ; Xiaojuan He,
| |
Collapse
|
20
|
Mirzadeh Azad F, Taheri Bajgan E, Naeli P, Rudov A, Bagheri Moghadam M, Sadat Akhtar M, Gholipour A, Mowla SJ, Malakootian M. Differential Expression Pattern of linc-ROR Spliced Variants in Pluripotent and Non-Pluripotent Cell Lines. CELL JOURNAL 2022; 24:569-576. [PMID: 36259474 PMCID: PMC9617025 DOI: 10.22074/cellj.2022.8205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The human large intergenic non-coding RNA-regulator of reprogramming program (linc-ROR) is known as a stem cell specific linc-RNA. linc-ROR counteracts differentiation via sequestering microRNA-145 (miR-145) that targets OCT4 transcript. Despite the research on the expression and function, the exact structure of linc-ROR transcripts is not clear. Considering the contribution of alternative splicing in transcripts structures and function, identifying different spliced variants of linc-ROR is necessary for further functional analyses. We aimed to find the alternatively spliced transcripts of linc-ROR and investigate their expression pattern in stem and cancer cell lines and during neural differentiation of NT2 cells as a model for understanding linc-ROR role in stem cell and differentiation. MATERIALS AND METHODS In this experimental study, linc-ROR locus was scanned for identifying novel exons. Different primer sets were used to detect new spliced variants by reverse transcription polymerase chain reaction (RT-PCR) and direct sequencing. Quantitative PCR (qPCR) and RT-PCR were employed to profile expression of linc-ROR transcripts in different cell lines and during neural differentiation of stem cells. RESULTS We could discover 13 novel spliced variants of linc-ROR harboring unique array of exons. Our work uncovered six novel exons, some of which were the product of exonized transposable elements. Monitoring expression profile of the linc-ROR spliced variants in a panel of pluripotent and non-pluripotent cells exhibited that all transcripts were primarily expressed in pluripotent cells. Moreover, the examined linc-ROR spliced variants showed a similar downregulation during neural differentiation of NT2 cells. CONCLUSION Altogether, our data showed despite the difference in the structure and composition of exons, various spliced variants of linc-ROR showed similar expression pattern in stem cells and through differentiation.
Collapse
Affiliation(s)
- Fatemeh Mirzadeh Azad
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran,Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Elham Taheri Bajgan
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parisa Naeli
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran,Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Alexander Rudov
- Department of Biomolecular Sciences, University of Urbino, Via Saffi Urbino, Italy
| | - Mahrokh Bagheri Moghadam
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mozhgan Sadat Akhtar
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akram Gholipour
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Javad Mowla
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran,Cardiogenetic Research CenterRajaie Cardiovascular Medical and Research CenterIran University
of Medical SciencesTehranIran
| |
Collapse
|
21
|
Zhang H, Song J, Zhou X. Long Noncoding RNA P53 Upregulated Regulator of P53 Levels Promotes Osteogenic Differentiation in Osteoporosis Progression Through Sponging miR-135a-5p. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study aimed to explore the expression of long noncoding RNA p53 upregulated regulator of P53 levels (lncRNA PURPL) and microRNA (miR)-135a-5p in osteoporosis and their role in osteogenic differentiation. The relationship between lncRNA PURPL and miR-135a-5p was confirmed by Star-Base
and luciferase reporter assay. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) assay was used to detect lncRNA PURPL and miR-135a-5p expression. RT-qPCR and western blot analysis were used to measure osteogenic markers expression. Alkaline phosphatase (ALP) activity
was also determined. Results indicated that lncRNA PURPL binds to miR-135a-5p. lncRNA PURPL expression was decreased and miR-135a-5p expression was increased in patients with osteoporosis. In the process of osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs), the
expression levels of osteoblast markers including RUNX family transcription factor 2 (Runx2), ALP and Osterix, and ALP activity were significantly increased. Besides, lncRNA PURPL was improved, while miR-135a-5p was down-regulated during the osteogenic differentiation of hBMSCs. Moreover,
lncRNA PURPL-siRNA significantly decreased the expression of ALP, Runx2 and Osterix, and reduced ALP activity in hBMSCs subjected to osteogenic induction, while all of these effects were reversed by miR-135a-5p inhibitor. In conclusion, lncRNA PURPL/miR-135a-5p may be a new axis for osteoporosis
treatment.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000, China
| | - Jie Song
- Department of Geriatrics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000, China
| | - Xianjie Zhou
- Department of Orthopedics, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huangshi, 435000, China
| |
Collapse
|
22
|
Wang J, Cui Y, Liu H, Li S, Sun S, Xu H, Peng C, Wang Y, Wu D. MicroRNA-loaded biomaterials for osteogenesis. Front Bioeng Biotechnol 2022; 10:952670. [PMID: 36199361 PMCID: PMC9527286 DOI: 10.3389/fbioe.2022.952670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
The large incidence of bone defects in clinical practice increases not only the demand for advanced bone transplantation techniques but also the development of bone substitute materials. A variety of emerging bone tissue engineering materials with osteogenic induction ability are promising strategies for the design of bone substitutes. MicroRNAs (miRNAs) are a class of non-coding RNAs that regulate intracellular protein expression by targeting the non-coding region of mRNA3′-UTR to play an important role in osteogenic differentiation. Several miRNA preparations have been used to promote the osteogenic differentiation of stem cells. Therefore, multiple functional bone tissue engineering materials using miRNA as an osteogenic factor have been developed and confirmed to have critical efficacy in promoting bone repair. In this review, osteogenic intracellular signaling pathways mediated by miRNAs are introduced in detail to provide a clear understanding for future clinical treatment. We summarized the biomaterials loaded with exogenous cells engineered by miRNAs and biomaterials directly carrying miRNAs acting on endogenous stem cells and discussed their advantages and disadvantages, providing a feasible method for promoting bone regeneration. Finally, we summarized the current research deficiencies and future research directions of the miRNA-functionalized scaffold. This review provides a summary of a variety of advanced miRNA delivery system design strategies that enhance bone regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dankai Wu
- *Correspondence: Yanbing Wang, ; Dankai Wu,
| |
Collapse
|
23
|
Yang Z, Feng L, Wang M, Li Y, Bai S, Lu X, Wang H, Zhang X, Wang Y, Lin S, Tortorella MD, Li G. Sesamin Promotes Osteoporotic Fracture Healing by Activating Chondrogenesis and Angiogenesis Pathways. Nutrients 2022; 14:2106. [PMID: 35631249 PMCID: PMC9147588 DOI: 10.3390/nu14102106] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/31/2022] Open
Abstract
Osteoporotic fracture has been regarded as one of the most common bone disorders in the aging society. The natural herb-derived small molecules were revealed as potential treatment approaches for osteoporotic fracture healing. Sesamin is a member of lignan family, which possesses estrogenic activity and plays a significant role in modulating bone homeostasis. Our previous study reported the promoting effect of sesamin on postmenopausal osteoporosis treatment. However, the role of sesamin in osteoporotic fracture healing has not been well studied yet. In this study, we further investigated the putative treatment effect of sesamin on osteoporotic fracture healing. Our study indicated that sesamin could activate bone morphogenetic protein 2 (BMP2) signaling pathway and further promotes in vitro chondrogenesis and angiogenesis activities. This promoting effect was abolished by the treatment of ERα inhibitor. In the osteoporotic bone fracture model, we demonstrated that sesamin markedly improves the callus formation and increases the cartilaginous area at the early-stage, as well as narrowing the fracture gap, and expands callus volume at the late-stage fracture healing site of the OVX mice femur. Furthermore, the angiogenesis at the osteoporotic fracture site was also significantly improved by sesamin treatment. In conclusion, our research illustrated the therapeutic potential and underlying regulation mechanisms of sesamin on osteoporotic fracture healing. Our studies shed light on developing herb-derived bioactive compounds as novel drugs for the treatment of osteoporotic fracture healing, especially for postmenopausal women with low estrogen level.
Collapse
Affiliation(s)
- Zhengmeng Yang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China; (L.F.); (Y.W.)
| | - Ming Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Yucong Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Shanshan Bai
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Xuan Lu
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Haixing Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Xiaoting Zhang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China; (L.F.); (Y.W.)
| | - Sien Lin
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| | - Micky D. Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China; (L.F.); (Y.W.)
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; (Z.Y.); (M.W.); (Y.L.); (S.B.); (X.L.); (H.W.); (X.Z.); (S.L.)
| |
Collapse
|
24
|
Crisafulli L, Ficara F. Micro-RNAs: A safety net to protect hematopoietic stem cell self-renewal. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1693. [PMID: 34532984 PMCID: PMC9285953 DOI: 10.1002/wrna.1693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 11/05/2022]
Abstract
The hematopoietic system is sustained over time by a small pool of hematopoietic stem cells (HSCs). They reside at the apex of a complex hierarchy composed of cells with progressively more restricted lineage potential, regenerative capacity, and with different proliferation characteristics. Like other somatic stem cells, HSCs are endowed with long-term self-renewal and multipotent differentiation ability, to sustain the high turnover of mature cells such as erythrocytes or granulocytes, and to rapidly respond to acute peripheral stresses including bleeding, infections, or inflammation. Maintenance of both attributes over time, and of the proper balance between these opposite features, is crucial to ensure the homeostasis of the hematopoietic system. Micro-RNAs (miRNAs) are short non-coding RNAs that regulate gene expression posttranscriptionally upon binding to specific mRNA targets. In the past 10 years they have emerged as important players for preserving the HSC pool by acting on several biological mechanisms, such as maintenance of the quiescent state while preserving proliferation ability, prevention of apoptosis, premature differentiation, lineage skewing, excessive expansion, or retention within the BM niche. miRNA-mediated posttranscriptional fine-tuning of all these processes constitutes a safety mechanism to protect HSCs, by complementing the action of transcription factors and of other regulators and avoiding unwanted expansion or aplasia. The current knowledge of miRNAs function in different aspects of HSC biology, including consequences of aberrant miRNA expression, will be reviewed; yet unsolved issues will be discussed. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Laura Crisafulli
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNRMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| | - Francesca Ficara
- UOS Milan Unit, Istituto di Ricerca Genetica e Biomedica (IRGB), CNRMilanItaly
- IRCCS Humanitas Research HospitalMilanItaly
| |
Collapse
|
25
|
Shi CJ, Li SY, Shen CH, Pan FF, Deng LQ, Fu WM, Wang JY, Zhang JF. Icariside II suppressed tumorigenesis by epigenetically regulating the circβ-catenin-Wnt/β-catenin axis in colorectal cancer. Bioorg Chem 2022; 124:105800. [DOI: 10.1016/j.bioorg.2022.105800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
|
26
|
Zhang FW, Peng LY, Shi CJ, Li JC, Pang FX, Fu WM, Pan XH, Zhang JF. Baicalein mediates the anti-tumor activity in Osteosarcoma through lncRNA-NEF driven Wnt/β-catenin signaling regulatory axis. J Orthop Translat 2022; 33:132-141. [PMID: 35330943 PMCID: PMC8919235 DOI: 10.1016/j.jot.2021.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 02/02/2023] Open
Abstract
Background Osteosarcoma (OS) is a common type of malignant bone tumor in adolescents with high risk of metastasis. However, the clinical management still remains unsatisfactory. Traditional Chinese medicine (TCM) has been widely considered as an alternative treatment, and their extracts have proved to possess great potential for drug discovery. Baicalein (BA), the active pharmaceutical ingredient of rhizoma coptidis, was proved to have anti-tumor properties in OS, but the mechanism remains poorly understood. Methods The potential anti-cancer effects on cell growth, cell cycle, apoptosis and migration were examined in OS cells. Moreover, the lncRNA-Neighboring Enhancer of FOXA2 (lncRNA-NEF) and Wnt/β-catenin signaling were detected by qPCR and Western blotting assays. The in vivo effect of GA on tumor growth was investigated using a xenograft mice model. Results In the present study, BA was found to significantly suppress tumor growth in vitro and in vivo. And it was also found to inhibit the invasion and metastasis as well. As for the mechanism investigation, lncRNA-NEF was obviously upregulated by BA in OS cells, and thus induced the inactivation of Wnt/β-catenin signaling. Moreover, lncRNA-NEF knockdown partially reversed the BA-induced anti-cancer activities; and successfully compensated the suppressive effect on Wnt/β-catenin signaling. We therefore suggested that BA induced the inactivation of Wnt/β-catenin signaling through promoting lncRNA-NEF expression. Conclusions In conclude, our results demonstrated that BA suppressed tumor growth and metastasis in vitro and in vivo through an lncRNA-NEF driven Wnt/β-catenin regulatory axis, in which lncRNA-NEF was upregulated by BA, and thus induced the inactivation of Wnt/β-catenin signaling. The Translational potential of this article The findings derived from this study validates the anti-cancer activity of BA in OS and provides a novel underlying mechanism, which suggest that BA may be a potential candidate to develop the effective drug for OS patients.
Collapse
Affiliation(s)
- Feng-wei Zhang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, PR China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Li-yang Peng
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, PR China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Chuan-Jian Shi
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 511458, PR China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Jian-chi Li
- Department of Orthopaedics and Traumatology, Shenzhen Tenth People's Hospital Affiliated to Jinan University, University of Chinese Academy of Sciences Shenzhen Hospital (Guangming), Shenzhen, Guangdong, 518106, PR China
| | - Feng-xiang Pang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, PR China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Wei-ming Fu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 511458, PR China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Xiao-hua Pan
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, PR China
- Corresponding author. Department of Orthopaedics , The Second School of Clinical Medicine, Southern Medical University, PR China.
| | - Jin-fang Zhang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, PR China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
- Corresponding author. Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| |
Collapse
|
27
|
De la Fuente-Hernandez MA, Sarabia-Sanchez MA, Melendez-Zajgla J, Maldonado-Lagunas V. Role of lncRNAs into Mesenchymal Stromal Cell Differentiation. Am J Physiol Cell Physiol 2022; 322:C421-C460. [PMID: 35080923 DOI: 10.1152/ajpcell.00364.2021] [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: 11/22/2022]
Abstract
Currently, findings support that 75% of the human genome is actively transcribed, but only 2% is translated into a protein, according to databases such as ENCODE (Encyclopedia of DNA Elements) [1]. The development of high-throughput sequencing technologies, computational methods for genome assembly and biological models have led to the realization of the importance of the previously unconsidered non-coding fraction of the genome. Along with this, noncoding RNAs have been shown to be epigenetic, transcriptional and post-transcriptional regulators in a large number of cellular processes [2]. Within the group of non-coding RNAs, lncRNAs represent a fascinating field of study, given the functional versatility in their mode of action on their molecular targets. In recent years, there has been an interest in learning about lncRNAs in MSC differentiation. The aim of this review is to address the signaling mechanisms where lncRNAs are involved, emphasizing their role in either stimulating or inhibiting the transition to differentiated cell. Specifically, the main types of MSC differentiation are discussed: myogenesis, osteogenesis, adipogenesis and chondrogenesis. The description of increasingly new lncRNAs reinforces their role as players in the well-studied field of MSC differentiation, allowing a step towards a better understanding of their biology and their potential application in the clinic.
Collapse
Affiliation(s)
- Marcela Angelica De la Fuente-Hernandez
- Facultad de Medicina, Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sanchez
- Facultad de Medicina, Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | |
Collapse
|
28
|
The management of bone defect using long non-coding RNA as a potential biomarker for regulating the osteogenic differentiation process. Mol Biol Rep 2022; 49:2443-2453. [PMID: 34973122 PMCID: PMC8863721 DOI: 10.1007/s11033-021-07013-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023]
Abstract
Tissue engineered bone brings hope to the treatment of bone defects, and the osteogenic differentiation of stem cells is the key link. Inducing osteogenic differentiation of stem cells may be a potential approach to promote bone regeneration. In recent years, lncRNA has been studied in the field increasingly, which is believed can regulate cell cycle, proliferation, metastasis, differentiation and immunity, participating in a variety of physiology and pathology processes. At present, it has been confirmed that certain lncRNAs regulate the osteogenesis of stem cells and take part in mediating signaling pathways including Wnt/β-catenin, MAPK, TGF-β/BMP, and Notch pathways. Here, we provided an overview of lncRNA, reviewed its researches in the osteogenic differentiation of stem cells, emphasized the importance of lncRNA in bone regeneration, and focused on the roles of lncRNA in signaling pathways, in order to make adequate preparations for applying lncRNA to bone tissue Engineering, letting it regulate the osteogenic differentiation of stem cells for bone regeneration.
Collapse
|
29
|
Chen W, Jiang W, Dong J, Wang J, Wang B. MiR-200b-3p induces the formation of insulin-producing cells from umbilical cord mesenchymal stem cells by targeting ZEB2. Crit Rev Eukaryot Gene Expr 2022; 32:33-46. [DOI: 10.1615/critreveukaryotgeneexpr.2022041822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
30
|
Yang Z, Feng L, Wang H, Li Y, Lo JHT, Zhang X, Lu X, Wang Y, Lin S, Tortorella MD, Li G. DANCR Mediates the Rescuing Effects of Sesamin on Postmenopausal Osteoporosis Treatment via Orchestrating Osteogenesis and Osteoclastogenesis. Nutrients 2021; 13:4455. [PMID: 34960006 PMCID: PMC8704418 DOI: 10.3390/nu13124455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 02/03/2023] Open
Abstract
As one of the leading causes of bone fracture in postmenopausal women and in older men, osteoporosis worldwide is attracting more attention in recent decades. Osteoporosis is a common disease mainly resulting from an imbalance of bone formation and bone resorption. Pharmaceutically active compounds that both activate osteogenesis, while repressing osteoclastogenesis hold the potential of being therapeutic medications for osteoporosis treatment. In the present study, sesamin, a bioactive ingredient derived from the seed of Sesamum Indicum, was screened out from a bioactive compound library and shown to exhibit dual-regulating functions on these two processes. Sesamin was demonstrated to promote osteogenesis by upregulating Wnt/β-catenin, while repressing osteoclastogenesis via downregulating NF-κB signaling . Furthermore, DANCR was found to be the key regulator in sesamin-mediated bone formation and resorption . In an ovariectomy (OVX)-induced osteoporotic mouse model, sesamin could rescue OVX-induced bone loss and impairment. The increased serum level of DANCR caused by OVX was also downregulated upon sesamin treatment. In conclusion, our results demonstrate that sesamin plays a dual-functional role in both osteogenesis activation and osteoclastogenesis de-activation in a DANCR-dependent manner, suggesting that it may be a possible medication candidate for osteoporotic patients with elevated DNACR expression levels.
Collapse
Affiliation(s)
- Zhengmeng Yang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Lu Feng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China; (L.F.); (Y.W.)
| | - Haixing Wang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Yucong Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Jessica Hiu Tung Lo
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Xiaoting Zhang
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Xuan Lu
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China; (L.F.); (Y.W.)
| | - Sien Lin
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| | - Micky D. Tortorella
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong, China; (L.F.); (Y.W.)
| | - Gang Li
- Stem Cells and Regenerative Medicine Laboratory, Department of Orthopaedics & Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hospital, Hong Kong, China; (Z.Y.); (H.W.); (Y.L.); (J.H.T.L.); (X.Z.); (X.L.); (S.L.)
| |
Collapse
|
31
|
Sidharthan DS, Abhinandan R, Balagangadharan K, Selvamurugan N. Advancements in nucleic acids-based techniques for bone regeneration. Biotechnol J 2021; 17:e2100570. [PMID: 34882984 DOI: 10.1002/biot.202100570] [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: 10/19/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022]
Abstract
The dynamic biology of bone involving an enormous magnitude of cellular interactions and signaling transduction provides ample biomolecular targets, which can be enhanced or repressed to mediate a rapid regeneration of the impaired bone tissue. The delivery of nucleic acids such as DNA and RNA can enhance the expression of osteogenic proteins. Members of the RNA interference pathway such as miRNA and siRNA can repress negative osteoblast differentiation regulators. Advances in nanomaterials have provided researchers with a plethora of delivery modules that can ensure proper transfection. Combining the nucleic acid carrying vectors with bone scaffolds has met with tremendous success in accomplishing bone formation. Recent years have witnessed the advent of CRISPR and DNA nanostructures in regenerative medicine. This review focuses on the delivery of nucleic acids and touches upon the prospect of CRISPR and DNA nanostructures for bone tissue engineering, emphasizing their potential in treating bone defects.
Collapse
Affiliation(s)
- Dharmaraj Saleth Sidharthan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Ranganathan Abhinandan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Kalimuthu Balagangadharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
32
|
Guo Y, Sun P, Guo W, Yin Q, Han J, Sheng S, Liang J, Dong Z. LncRNA DDX11 antisense RNA 1 promotes EMT process of esophageal squamous cell carcinoma by sponging miR-30d-5p to regulate SNAI1/ZEB2 expression and Wnt/β-catenin pathway. Bioengineered 2021; 12:11425-11440. [PMID: 34866524 PMCID: PMC8810181 DOI: 10.1080/21655979.2021.2008759] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 12/11/2022] Open
Abstract
LncRNA DDX11 antisense RNA 1 (DDX11-AS1) is recognized as having an imperative oncogenic role in different types of human cancer. Nevertheless, the functions, as well as the basic mechanisms of DDX11-AS1 in the EMT process of esophageal squamous cell carcinoma (ESCC), are yet to be clarified. In this research, high DDX11-AS1 expression was detected in ESCC cells as well as tissues and was linked to the poor prognosis of patients with ESCC. DDX11-AS1 promoted cell proliferation, migration, invasion ability and epithelial mesenchymal transition (EMT) process in vitro. Mechanistic analysis depicted that DDX11-AS1 may function as a ceRNA through sponging miR-30d-5p to upregulate the expression of SNAI1 and ZEB2. Meanwhile, overexpression of DDX11-AS1 might cause the activation of the Wnt/β-catenin signaling pathway via targeting miR-30d-5p. On the whole, the findings of this research illustrate that DDX11-AS1 may act as an EMT-related lncRNA to advance ESCC progression through sponging miR-30d-5p to regulate SNAI1/ZEB2 expression and activate the Wnt/β-catenin pathway, which indicates that it might serve as a probable therapeutic target for ESCC.
Collapse
Affiliation(s)
- Yanli Guo
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pingping Sun
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wei Guo
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qing Yin
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Junshu Han
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Supeng Sheng
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jia Liang
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhiming Dong
- Hebei Cancer Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
33
|
Li J, Lu L, Liu Y, Yu X. Bone marrow adiposity during pathologic bone loss: molecular mechanisms underlying the cellular events. J Mol Med (Berl) 2021; 100:167-183. [PMID: 34751809 DOI: 10.1007/s00109-021-02164-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/09/2021] [Accepted: 11/03/2021] [Indexed: 02/05/2023]
Abstract
Bone marrow (BM) is a heterogeneous niche where bone marrow stromal cells (BMSCs), osteoblasts, osteoclasts, adipocytes, hematopoietic cells, and immune cells coexist. The cellular composition of BM changes with various pathophysiological states. A reduction in osteoblast number and a concomitant increase in adipocyte number in aging and pathological conditions put bone marrow adipose tissue (BMAT) into spotlight. Accumulating evidence strongly supports that an overwhelming production of BMAT is a major contributor to bone loss disorders. Therefore, BMAT-targeted therapy can be an efficient and feasible intervention for osteoporosis. However, compared to blocking bone-destroying molecules produced by BMAT, suppressing BMAT formation is theoretically a more effective and fundamental approach in treating osteoporotic bone diseases. Thus, a deep insight into the molecular basis underlying increased BM adiposity during pathologic bone loss is critical to formulate strategies for therapeutically manipulating BMAT. In this review, we comprehensively summarize the molecular mechanisms involved in adipocyte differentiation of BMSCs as well as the interaction between bone marrow adipocytes and osteoclasts. More importantly, we further discuss the potential clinical implications of therapeutically targeting the upstream of BMAT formation in bone loss diseases.
Collapse
Affiliation(s)
- Jiao Li
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Lingyun Lu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xijie Yu
- Department of Endocrinology and Metabolism, Laboratory of Endocrinology and Metabolism, Rare Disease Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan Province, China
| |
Collapse
|
34
|
Tang W, Liu Q, Tan W, Sun T, Deng Y. LncRNA expression profile analysis of Mg 2+-induced osteogenesis by RNA-seq and bioinformatics. Genes Genomics 2021; 43:1247-1257. [PMID: 34427873 DOI: 10.1007/s13258-021-01140-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND In recent years, magnesium (Mg) has been extensively studied for manufacturing biodegradable orthopedic devices. Besides other advantages, researches have shown that magnesium-based implants can stimulate osteogenesis thus accelerating orthopedic trauma recovery, but its molecular mechanism is not fully understood. Meanwhile, long non-coding RNA (lncRNA) has been found to play vital role in regulating osteogenic differentiation. OBJECTIVE To explore the role of lncRNA in Mg2+ (magnesium ions)-induced osteogenesis. METHODS The effect of Mg2+ on mBMSCs proliferation was detected by the CCK-8 assay. The optimum concentration of Mg2+ (7.5 mM) in promoting mBMSCs osteogenesis was determined by ALP staining and Alizarin red staining, western blot and RT-qPCR were performed to detect osteogenic markers expressions. The lncRNAs and mRNAs expression profiles of mBMSCs were assessed by RNA-Seq and processed by bioinformatics analysis. The selected lncRNAs expression level was validated by RT-qPCR. RESULTS The effect of Mg2+ in promoting osteogenesis was confirmed and the optimum concentration was determined as 7.5 mM. The lncRNAs and mRNAs differentially expressed between 7.5 mM Mg2+-treated group and control group was detected and functional analysis revealed that their function were associated with osteogenesis. The ceRNA networks were constructed for H19 and Dubr that aberrantly expressed in two groups. The ceRNA networks of selected lncRNAs (H19 and Dubr) were constructed. CONCLUSIONS This study identified H19 and Dubr as osteogenic associated lncRNAs involved in Mg2+-induced osteogenesis, and they might play their roles through lncRNA-miRNA-mRNA axis.
Collapse
Affiliation(s)
- Wen Tang
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Qing Liu
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Wei Tan
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Tianshi Sun
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Youwen Deng
- Department of Spine Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
35
|
Taheri Bajgan E, Gholipour A, Faghihi M, Mowla SJ, Malakootian M. Linc-ROR has a Potential ceRNA Activity for OCT4A by Sequestering miR-335-5p in the HEK293T Cell Line. Biochem Genet 2021; 60:1007-1024. [PMID: 34669056 DOI: 10.1007/s10528-021-10140-0] [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: 06/10/2021] [Accepted: 10/07/2021] [Indexed: 12/26/2022]
Abstract
Linc-ROR has a regulatory role in reprogramming, and the core stem cell transcription factors, OCT4, SOX2, and NANOG, regulate its expression. MicroRNAs (miRNAs) are also a critical constituent of pivotal posttranscriptional regulatory pathways. One of such interactions is a competing endogenous RNA interaction that connects small and long non-coding RNAs with coding transcripts. Here, we aimed to investigate the existence of such associations between OCT4A, Linc-ROR, hsa-miR-335-5p, and hsa-miR-544. Bioinformatic analysis was performed to evaluate the expression status of OCT4A, Linc-ROR, miR-335, and miR-544 throughout differentiation as well as in various differentiated cells. The complete lengths of OCT4A and Linc-ROR, and OCT4A 3'-UTR were cloned in the luciferase reporter vector, and the precursors of miR-335 and miR-544 were cloned in expression vectors. Following the overexpression of miR-335 and miR-544 in the 5637 cell line, the endogenous expression of OCT4A and Linc-ROR was evaluated. Afterward, the expression vectors of miRNAs and the reporter vectors of OCT4A/Linc-ROR were co-transfected in the HEK293T cell line. Via the Dual-Luciferase assay, the effect of the overexpression of miRNAs on their two possible targets (Linc-ROR and OCT4A) was investigated. The bioinformatic analysis demonstrated a relatively similar expression pattern for OCT4A and Linc-ROR, while miR-335 showed a different expression status. Both miR-335 and miR-544 inhibited the endogenous expression of OCT4A. The Dual-Luciferase assay likewise confirmed the inhibitory effect of miR-335 and miR-544 on OCT4A expression. In contrast, the miR-335 inhibitory effect was reversed in the presence of Linc-ROR, resulting in the upregulation of OCT4A. Such evidence suggests that Linc-ROR may compete with OCT4A to interact with miR-335.
Collapse
Affiliation(s)
- Elham Taheri Bajgan
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Akram Gholipour
- Department of Biology, Islamic Azad University Tehran Science and Research Branch, Tehran, Iran
| | - Mohammadali Faghihi
- Center for Therapeutic Innovation and Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL, 33136, USA
| | - Seyed Javad Mowla
- Molecular Genetics Department, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahshid Malakootian
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Fu Y, Hu X, Gao Y, Li K, Fu Q, Liu Q, Liu D, Zhang Z, Qiao J. LncRNA ROR/miR-145-5p axis modulates the osteoblasts proliferation and apoptosis in osteoporosis. Bioengineered 2021; 12:7714-7723. [PMID: 34617877 PMCID: PMC8806809 DOI: 10.1080/21655979.2021.1982323] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Osteoporosis (OP) is a systemic bone metabolic disease. Promotion of osteoblast proliferation and inhibition of cell apoptosis may be helpful for the prevention and clinical treatment of OP. In the current study, we focused on the expression changes and clinical values of lncRNA ROR and miR-145-5p in OP clinical serum samples, and investigated the interactive modulation effect of ROR/miR-145-5p on osteoblast function. Serum samples were obtained from 82 OP patients and 79 healthy individuals. MC3T3-E1 was applied for the cell experiments. Levels of lncRNA ROR and miR-145-5p were detected using qRT-PCR. Transient transfection was performed to regulate gene levels in cells, and cell proliferation and apoptosis were detected. A reciprocal correlation between lncRNA ROR and miR-145-5p was explored. LncRNA ROR was downregulated, and miR-145-5p was overexpressed in OP patients. The combined diagnosis of ROR and miR-145-5p showed good diagnostic value for OP. ROR knockdown promoted the MC3T3-E1 cell apoptosis and inhibited cell proliferation. Luciferase reporting assay verified the target relationship between ROR and miR-145-5p. MiR-145-5p downregulation reversed ROR silence mediated effect on MC3T3-E1 cell proliferation and apoptosis. LncRNA ROR is downregulated and miR-145-5p is highly expressed in OP patients. ROR knockdown may inhibit osteoblast proliferation via targeting miR-145-5p. It may provide a theoretical basis and experimental basis for ROR to be a potential target for the treatment of OP.
Collapse
Affiliation(s)
- Yin Fu
- Department of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyang Hu
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yanyu Gao
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Kai Li
- Department of Interventional Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qiang Fu
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingpeng Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dan Liu
- Brown University RI Hospital Liver Research Center, Providence, RI, USA
| | - Zhijia Zhang
- Beijing University of Chinese Medicine, Beijing, China
| | - Jiutao Qiao
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
37
|
Yang X, Shao J, Wu XM, Pan FF, Yang SA, Pan XH, Jin AM. Troxerutin Stimulates Osteoblast Differentiation of Mesenchymal Stem Cell and Facilitates Bone Fracture Healing. Front Pharmacol 2021; 12:723145. [PMID: 34434113 PMCID: PMC8381475 DOI: 10.3389/fphar.2021.723145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/29/2021] [Indexed: 11/23/2022] Open
Abstract
Troxerutin (TRX), a semi-synthetic derivative of the natural bioflavonoid rutin, is a bioactive flavonoid widely abundant in various fruits and vegetables. Known as vitamin P4, TRX has been demonstrated to have several activities including anti-inflammation, anti-oxidants, vasoprotection, and immune support in various studies. Although rutin, the precursor of troxerutin, was reported to have a protective role against bone loss, the function of TRX in skeletal system remains unknown. In the present study, we found that TRX promoted osteogenic differentiation of human mesenchymal stem cells (MSCs) in a concentration-dependent manner by stimulating the alkaline phosphatase (ALP) activity, calcium nodule formation and osteogenic marker genes expression in vitro. The further investigation demonstrated that TRX stimulated the expression of the critical transcription factor β-catenin and several downstream target genes of Wnt signaling, thus activated Wnt/β-catenin signaling. Using a femur fracture rats model, TRX was found to stimulate new bone formation and accelerate the fracture healing in vivo. Collectively, our data demonstrated that TRX could promote osteogenesis in vitro and facilitate the fracture healing in vivo, indicating that TRX may be a promising therapeutic candidate for bone fracture repair.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Shao
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-Min Wu
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, China
| | - Fei-Fei Pan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shao-An Yang
- Department of Traumatic Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao-Hua Pan
- Department of Orthopaedics, The Second School of Clinical Medicine, Southern Medical University, The Second Affiliated Hospital of Shenzhen University, The Clinical Medical College of Guangdong Medical University, People's Hospital of Shenzhen Baoan District, Shenzhen, China
| | - An-Min Jin
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
38
|
Wang X, He J, Wang H, Zhao D, Geng B, Wang S, An J, Wang C, Han H, Xia Y. Fluid shear stress regulates osteoblast proliferation and apoptosis via the lncRNA TUG1/miR-34a/FGFR1 axis. J Cell Mol Med 2021; 25:8734-8747. [PMID: 34350720 PMCID: PMC8435422 DOI: 10.1111/jcmm.16829] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/11/2021] [Accepted: 07/15/2021] [Indexed: 01/22/2023] Open
Abstract
LncRNAs and microRNAs play critical roles in osteoblast differentiation and bone formation. However, their exact roles in osteoblasts under fluid shear stress (FSS) and the possible mechanisms remain unclear. The aim of this study was to explore whether and how miR‐34a regulates osteoblast proliferation and apoptosis under FSS. In this study, FSS down‐regulated miR‐34a levels of MC3T3‐E1 cells. MiR‐34a up‐regulation attenuated FSS‐induced promotion of proliferation and suppression of apoptosis. Luciferase reporter assay revealed that miR‐34a directly targeted FGFR1. Moreover, miR‐34a regulated osteoblast proliferation and apoptosis via FGFR1. Further, we validated that lncRNA TUG1 acted as a competing endogenous RNA (ceRNA) to interact with miR‐34a and up‐regulate FGFR1 protein expression. Furthermore, lncRNA TUG1 could promote proliferation and inhibit apoptosis. Taken together, our study revealed the key role of the lncRNA TUG1/miR‐34a/FGFR1 axis in FSS‐regulated osteoblast proliferation and apoptosis and may provide potential therapeutic targets for osteoporosis.
Collapse
Affiliation(s)
- Xingwen Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Jinwen He
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Hong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Dacheng Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Shenghong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Jiangdong An
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Cuifang Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Hua Han
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou, Gansu, China.,Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
39
|
Zhu Y, You J, Wei W, Gu J, Xu C, Gu X. Downregulated lncRNA RCPCD promotes differentiation of embryonic stem cells into cardiac pacemaker-like cells by suppressing HCN4 promoter methylation. Cell Death Dis 2021; 12:667. [PMID: 34215719 PMCID: PMC8253811 DOI: 10.1038/s41419-021-03949-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/25/2022]
Abstract
Long non-coding RNA (lncRNA) is receiving increasing attention in embryonic stem cells (ESCs) research. However, the roles of lncRNA in the differentiation of ESCs into pacemaker-like cells are still unclear. Therefore, the present study aims to explore the roles and mechanisms of lncRNA in the differentiation of ESCs into pacemaker-like cells. ESCs were cultured and induced differentiation to pacemaker-like cells. RNA sequencing was used to identify the differential expression lncRNAs during the differentiation of ESCs into pacemaker-like cells. Cell morphology observation, flow cytometry, quantitative real-time polymerase chain reaction, western blot, and immunofluorescence were used to detect the differentiation of ESCs into pacemaker-like cells. LncRNA and genes overexpression or knockdown through transfected adenovirus in the differentiation process. The fluorescence in situ hybridization (FISH) detected the lncRNA location in the differentiated ESCs. Luciferase reporter gene assay, methylation-specific PCR, chromatin immunoprecipitation assay, and RNA immunoprecipitation assay were performed to reveal the mechanism of lncRNA-regulating HCN4 expression. Rescue experiments were used to confirm that lncRNA regulates the differentiation of ESCs into pacemaker-like cells through HCN4. We cultured the ESCs and induced the differentiation of ESCs into pacemaker-like cells successfully. The expression of lncRNA RCPCD was significantly decreased in the differentiation of ESCs into pacemaker-like cells. Overexpression of RCPCD inhibited the differentiation of ESCs into pacemaker-like cells. RCPCD inhibited the expression of HCN4 by increasing HCN4 methylation at the promoter region through DNMT1, DNMT2, and DNMT3. RCPCD inhibited the differentiation of ESCs into pacemaker-like cells by inhibiting the expression of HCN4. Our results confirm the roles and mechanism of lncRNA RCPCD in the differentiation of ESCs into pacemaker-like cells, which could pave the path for the development of a cell-based biological pacemaker.
Collapse
Affiliation(s)
- Ye Zhu
- Clinical Medical College of Yangzhou University, Yangzhou, China. .,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China.
| | - Jia You
- Department of Internal Medicine, Yangzhou Maternal and Child Health Care Hospital, Yangzhou, Jiangsu, 225001, China
| | - Wei Wei
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jianjun Gu
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Science Center, Oklahoma City, OK, 73104, US
| | - Xiang Gu
- Clinical Medical College of Yangzhou University, Yangzhou, China.,Department of Cardiology, Northern Jiangsu People's Hospital, Yangzhou, China
| |
Collapse
|
40
|
He Q, Li R, Hu B, Li X, Wu Y, Sun P, Jia Y, Guo Y. Stromal cell-derived factor-1 promotes osteoblastic differentiation of human bone marrow mesenchymal stem cells via the lncRNA-H19/miR-214-5p/BMP2 axis. J Gene Med 2021; 23:e3366. [PMID: 34032330 DOI: 10.1002/jgm.3366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Stromal cell-derived factor-1 (SDF-1) plays an important role in the osteoblastic differentiation of human bone marrow mesenchymal stem cells (hBMMSCs), but the specific mechanism remains unclear. Our study aimed to clarify the role of the lncRNA-H19/miR-214-5p/BMP2 axis in the osteoblastic differentiation of hBMMSCs induced by SDF-1. METHODS We used reverse-transcriptase polymerase chain reaction, western blotting, alkaline phosphatase activity test, and Alizarin red staining to evaluate the osteoblastic differentiation of primary hBMMSCs and the luciferase reporter assay to determine if lncRNA-H19 binds with miR-214-5p. RESULTS Our results indicated that SDF-1 (50 ng/mL) promotes the osteoblastic differentiation of hBMMSCs, significantly upregulates osteoblastogenic genes (OCN, OSX, RUNX2, and ALP), and increases Alizarin red staining, alkaline phosphatase activity, and lncRNA-H19 expression. Luciferase reporter assay verified that lncRNA-H19 binds with and represses miR-214-5p, thereby upregulating BMP2 expression. Use of miR-214-5p inhibitor or overexpression of lncRNA-H19 can promote the osteoblastic differentiation of hBMMSCs, but miR-214-5p or shH19 inhibits the osteoblastic differentiation of hBMMSCs. Treatment with an miR-214-5p inhibitor could rescue the inhibitory effect of shH19 on the osteoblastic differentiation of hBMMSCs. CONCLUSIONS Taken together, SDF-1 promotes the osteoblastic differentiation of hBMMSCs through the lncRNA-H19/miR-214-5p/BMP2 axis. Increased osteoblastic differentiation by an miR-214-5p inhibitor reveals a new possible strategy for the treatment of bone defect and osteoporosis.
Collapse
Affiliation(s)
- Qiting He
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ruibin Li
- Department of Orthopedic Surgery, Linyi central hospital, Linyi, Shandong, China
| | - Beibei Hu
- Department of Ultrasound, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xuezhou Li
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yunpeng Wu
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Pengfei Sun
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yuhua Jia
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yongyuan Guo
- Department of Orthopedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
41
|
Mishra P, Kumar S. Association of lncRNA with regulatory molecular factors in brain and their role in the pathophysiology of schizophrenia. Metab Brain Dis 2021; 36:849-858. [PMID: 33608830 DOI: 10.1007/s11011-021-00692-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/11/2021] [Indexed: 01/12/2023]
Abstract
Schizophrenia is one of the most agonizing neurodegenerative diseases of the brain. Research undertaken to understand the molecular mechanism of this disease has undergone a transition and currently more emphasis is put on long noncoding RNA (lncRNA). High expression level of lncRNA in the brain contributes to several molecular pathways essential for the proper functioning of neurons, neurotransmitters, and synapses, that are often found dysfunctional in Schizophrenia. Recently, the association of lncRNA with various molecular factors in the brain has been explored to a considerably large extent. This review comprehends the significance of lncRNA in causing profound regulatory effect in the brain and how any alterations to the association of lncRNA with regulatory proteins, enzymes and other noncoding RNA could contribute to the aetiology of Schizophrenia.
Collapse
Affiliation(s)
- Parinita Mishra
- Life Science Department, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Santosh Kumar
- Life Science Department, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
42
|
Zhang C, Zuo Q, Gao X, Hu C, Zhou S, Chen C, Zou Y, Zhao J, Zhang Y, Li B. H3K4me2 Promotes the Activation of lncCPSET1 by Jun in the Chicken PGC Formation. Animals (Basel) 2021; 11:1572. [PMID: 34072197 PMCID: PMC8227976 DOI: 10.3390/ani11061572] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Primordial germ cells are the ancestors of female and male cells. Current research has shown that long non-coding RNA (lncRNA) and Histone methylation are the pivotal epigenetic factors in the PGC formation. However, there are few studies on the regulatory mechanism of lncRNA in the formation of PGC. Here, we define the lncRNA highly expressed in chicken PGC, lncCPSET1 (chicken-PGC-specifically-expressed transcript 1) This study found that compared with the interference of lncCPSET1/histone methylase Mll2 alone, the PGC formation was severely inhibited with the interference of lncCPSET1 and histone methylase Mll2 jointly in vivo and in vitro. Studies on the transcription level of lncCPSET1 found that H3K4me2 and transcription factor Jun have a positive effect on the activation of lncCPSET1; while DNA hypomethylation inhibits the expression of lncCPSET1. In terms of mechanism, compared with DNA methylation, H3K4me2 dominates lncCPSET1 activation. H3K4me2 can be enriched in the lncCPSET1 promoter, change its chromosome conformation, recruit the transcription factor Jun, and activate the expression of lncCPSET1. Taken together, we confirmed the model that H3K4me2 rather than DNA hypomethylation mediates Jun to regulate lncCPSET1 transcription, which broadens the study of lncCPSET1 pre-transcriptional mechanism.
Collapse
Affiliation(s)
- Chen Zhang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Xiaomin Gao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Cai Hu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Shujian Zhou
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Chen Chen
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Yichen Zou
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Juanjuan Zhao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Yani Zhang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (C.Z.); (Q.Z.); (X.G.); (C.H.); (S.Z.); (C.C.); (Y.Z.); (J.Z.); (Y.Z.)
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212000, China
| |
Collapse
|
43
|
Wang H, Cao X, Wen X, Li D, Ouyang Y, Bao B, Zhong Y, Qin Z, Yin M, Chen Z, Yin X. Transforming growth factor‑β1 functions as a competitive endogenous RNA that ameliorates intracranial hemorrhage injury by sponging microRNA‑93‑5p. Mol Med Rep 2021; 24:499. [PMID: 33955515 PMCID: PMC8127068 DOI: 10.3892/mmr.2021.12138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/08/2021] [Indexed: 11/26/2022] Open
Abstract
Intracerebral hemorrhage (ICH) has the highest mortality rate of all stroke subtypes but an effective treatment has yet to be clinically implemented. Transforming growth factor-β1 (TGF-β1) has been reported to modulate microglia-mediated neuroinflammation after ICH and promote functional recovery; however, the underlying mechanisms remain unclear. Non-coding RNAs such as microRNAs (miRNAs) and competitive endogenous RNAs (ceRNAs) have surfaced as critical regulators in human disease. A known miR-93 target, nuclear factor erythroid 2-related factor 2 (Nrf2), has been shown to be neuroprotective after ICH. It was hypothesized that TGF-β1 functions as a ceRNA that sponges miR-93-5p and thereby ameliorates ICH injury in the brain. Short interfering RNA (siRNA) was used to knock down TGF-β1 and miR-93 expression was also pharmacologically manipulated to elucidate the mechanistic association between miR-93-5p, Nrf2, and TGF-β1 in an in vitro model of ICH (thrombin-treated human microglial HMO6 cells). Bioinformatics predictive analyses showed that miR-93-5p could bind to both TGF-β1 and Nrf2. It was found that neuronal miR-93-5p was dramatically decreased in these HMO6 cells, and similar changes were observed in fresh brain tissue from patients with ICH. Most importantly, luciferase reporter assays were used to demonstrate that miR-93-5p directly targeted Nrf2 to inhibit its expression and the addition of the TGF-β1 untranslated region restored the levels of Nrf2. Moreover, an miR-93-5p inhibitor increased the expression of TGF-β1 and Nrf2 and decreased apoptosis. Collectively, these results identified a novel function of TGF-β1 as a ceRNA that sponges miR-93-5p to increase the expression of neuroprotective Nrf2 and decrease cell death after ICH. The present findings provided evidence to support miR-93-5p as a potential therapeutic target for the treatment of ICH.
Collapse
Affiliation(s)
- Han Wang
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xianming Cao
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaoqing Wen
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Dongling Li
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Yetong Ouyang
- Department of Neurology, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi 330006, P.R. China
| | - Bing Bao
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Yuqin Zhong
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhengfang Qin
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Min Yin
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiying Chen
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiaoping Yin
- Department of Neurology, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, P.R. China
| |
Collapse
|
44
|
Zhang Y, Liu MW, He Y, Deng N, Chen Y, Huang J, Xie W. Protective effect of resveratrol on estrogen deficiency-induced osteoporosis though attenuating NADPH oxidase 4/nuclear factor kappa B pathway by increasing miR-92b-3p expression. Int J Immunopathol Pharmacol 2021; 34:2058738420941762. [PMID: 32674689 PMCID: PMC7370339 DOI: 10.1177/2058738420941762] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Introduction: Resveratrol (RES) exhibits estrogen-like effects and has potential applications to treatment of osteoporosis caused by estrogen deficiency; however, the specific mechanism of action of RES remains unclear. Here, we examined the therapeutic effects of RES on ovariectomized (OVX) rats with osteoporosis and determined the underlying mechanism. Methods: We established an OVX rat model to study osteoporosis caused by estrogen deficiency. The treatment groups were given orally with RES (50, 100, and 200 mg/day), the estrogen group received 0.8 mg/kg E2 daily via oral route, and the sham-operated and control groups received an equivalent dose of sodium carboxymethylcellulose orally. After 12 weeks of treatment, we used real-time quantitative polymerase chain reaction (PCR) and Western blot analysis to measure the gene and protein expression of miR-92b-3p, Nox4, NF-κBp65, IκB, BMP2, Smad7, and RUNX-2 in bone tissues. Right femur structural parameters were evaluated by micro-CT. Dual-energy X-ray 4500 W was used to determine systemic bone mineral density (BMD). Enzyme-linked immunosorbent assay (ELISA) kits were used to determine the serum levels of bone alkaline phosphatase (BALP), osteoprotegerin (OPG), anti-tartrate acid phosphatase-5b (PTRA5b), and carboxylated terminal peptide (CTX-I). The rat femoral bone specimens were stained using hematoxylin and eosin for pathological examination Results: We observed increased levels of serum estrogen in both ovaries, elevated miR-92b-3p levels in bone tissues, reduced levels of Nox4, NF-κBp65, p-IκB-a, and cathepsin K, and elevated gene and protein expression of BMP2, Smad7, and RUNX-2 in the OVX rat model of osteoporosis after treatment with RES. Elevated levels of BALP, OPG, ALP, and BMD along with reduced levels of TRAP-5b and CTX-I were also observed. The structural model index (SMI) and the trabecular space (Tb. Sp) decreased, while the trabecular thickness (Tb. Th), bone volume fraction (BV/TV), trabecular number (Tb.N), and tissue bone density (Conn.D) increased, thereby improving osteoporosis induced by estrogen deficiency in both ovaries. Conclusion: Cathepsin K expression and Nox4/NF-κB signaling pathway were suppressed by the elevated expression of miR-92b-3p. This inhibition was pivotal in the protective effect of RES against osteoporosis induced by estrogen deficiency in both ovaries. Thus, RES efficiently alleviated osteoporosis induced by estrogen deficiency in rats.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Traditional Chinese Medicine, The Third People's Hospital of Yunnan Province, Kunming, China
| | - Ming-Wei Liu
- Department of Emergency Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yun He
- Department of Orthopedics, Calmett Hospital & The First Hospital of Kunming, Kunming, China
| | - Ning Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yan Chen
- Normal Human Anatomy and Histological Embryology Department, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Jiecong Huang
- Department of Encephalopathy, Guangzhou Conghua Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Wei Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Wang H, Zhang K, Liu J, Yang J, Tian Y, Yang C, Li Y, Shao M, Su W, Song N. Curcumin Regulates Cancer Progression: Focus on ncRNAs and Molecular Signaling Pathways. Front Oncol 2021; 11:660712. [PMID: 33912467 PMCID: PMC8072122 DOI: 10.3389/fonc.2021.660712] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
Curcumin [(1E,6E) ‑1,7‑bis(4‑hydroxy‑3‑methoxyphenyl) hepta‑1,6‑diene‑3,5‑ dione] is a natural polyphenol derived from the rhizome of the turmeric plant Curcuma longa. Accumulated evidences have presented curcumin’s function in terms of anti-inflammatory, antioxidant properties, and especially anti-tumor activities. Studies demonstrated that curcumin could exert anti-tumor activity via multiple biological signaling pathways, such as PI3K/Akt, JAK/STAT, MAPK, Wnt/β-catenin, p53, NF-ĸB and apoptosis related signaling pathways. Moreover, Curcumin can inhibit tumor proliferation, angiogenesis, epithelial-mesenchymal transition (EMT), invasion and metastasis by regulating tumor related non-coding RNA (ncRNA) expression. In this review, we summarized the roles of curcumin in regulating signaling pathways and ncRNAs in different kinds of cancers. We also discussed the regulatory effect of curcumin through inhibiting carcinogenic miRNA and up regulating tumor suppressive miRNA. Furthermore, we aim to illustrate the cross regulatory relationship between ncRNA and signaling pathways, further to get a better understanding of the anti-tumor mechanism of curcumin, thus lay a theoretical foundation for the clinical application of curcumin in the future.
Collapse
Affiliation(s)
- Haijun Wang
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.,School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ke Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jia Liu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jie Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yidan Tian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Chen Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yushan Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Minglong Shao
- Department of Mental Health, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wei Su
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Na Song
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.,Institute of Precision Medicine, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
46
|
Jiang B, Zhu H, Tang L, Gao T, Zhou Y, Gong F, Tan Y, Xie L, Wu X, Li Y. Apatinib Inhibits Stem Properties and Malignant Biological Behaviors of Breast Cancer Stem Cells by Blocking Wnt/β-catenin Signal Pathway Through Down-regulating LncRNA ROR. Anticancer Agents Med Chem 2021; 22:1723-1734. [PMID: 33845750 DOI: 10.2174/1871520621666210412103849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/01/2021] [Accepted: 02/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer stem cells could influence tumor recurrence and metastasis. OBJECTIVE To develop a new effective treatment modality targeting breast cancer stem cells (BCSCs), and to explore the role of Apatinib in BCSCs. METHODS BCSCs were isolated from MDA-MB-231 cells by immune magnetic beads method. BCSCs were treated with Apatinib, lentiviral plasmids (lncRNA ROR) and iCRT-3 (Wnt pathway inhibitors). Viability, colony numbers, sphere numbers, apoptosis, migration, invasion of BCSCs were detected by MTT, colony formation, tumor sphere, flow cytometry, wound-healing, transwell assays, respectively. The expressions of markers (ABCG2, CD44, CD90, and CD24), epithelial-mesenchymal transition (EMT)-related molecules (E-cadherin, N-cadherin, Vimentin, MMP-2, MMP-9), and Wnt/β-catenin pathway-related proteins (Wnt3a, Wnt5a, β-catenin) in breast cancer stem cells were determined by performing Western blot and qRT-PCR analysis. RESULTS Apatinib decreased the viability and colony numbers of BCSCs in a concentration-dependent manner, and it also reduced sphere numbers, suppressed migration, invasion and lncRNA ROR expression, and induced apoptosis of BCSCs. However, these results were partially reversed by lncRNA ROR overexpression. Apatinib suppressed stem property, EMT process and Wnt/β-catenin pathway in BCSCs, which was partially reversed by lncRNA ROR overexpression. Moreover, lncRNA ROR overexpression increased the colony and sphere numbers, and promoted the cell viability, apoptosis inhibition, migration and invasion of BCSCs, but these effects were partially reversed by iCRT-3. LncRNA ROR overexpression increased the stem property, EMT process and Wnt/β-catenin pathway, which were partially counteracted by iCRT-3. CONCLUSION Apatinib inhibited stem property and malignant biological behaviors of BCSCs by blocking Wnt/β-catenin signal pathway through down-regulating lncRNA ROR.
Collapse
Affiliation(s)
- Baohong Jiang
- Department of Pharmacy, The First Affiliated Hospital, University of South China. China
| | - Hongbo Zhu
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Liting Tang
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Ting Gao
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Yu Zhou
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Fuqiang Gong
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Yeru Tan
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Liming Xie
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Xiaoping Wu
- Department of Medical Oncology, The First Affiliated Hospital, University of South China. China
| | - Yuehua Li
- Department of Medical Oncology, The First Affiliated Hospital, University of South China, No.69, Chuanshan Road, Hengyang, Hunan Province, 421001. China
| |
Collapse
|
47
|
Lanzillotti C, De Mattei M, Mazziotta C, Taraballi F, Rotondo JC, Tognon M, Martini F. Long Non-coding RNAs and MicroRNAs Interplay in Osteogenic Differentiation of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:646032. [PMID: 33898434 PMCID: PMC8063120 DOI: 10.3389/fcell.2021.646032] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/11/2021] [Indexed: 12/23/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have gained great attention as epigenetic regulators of gene expression in many tissues. Increasing evidence indicates that lncRNAs, together with microRNAs (miRNAs), play a pivotal role in osteogenesis. While miRNA action mechanism relies mainly on miRNA-mRNA interaction, resulting in suppressed expression, lncRNAs affect mRNA functionality through different activities, including interaction with miRNAs. Recent advances in RNA sequencing technology have improved knowledge into the molecular pathways regulated by the interaction of lncRNAs and miRNAs. This review reports on the recent knowledge of lncRNAs and miRNAs roles as key regulators of osteogenic differentiation. Specifically, we described herein the recent discoveries on lncRNA-miRNA crosstalk during the osteogenic differentiation of mesenchymal stem cells (MSCs) derived from bone marrow (BM), as well as from different other anatomical regions. The deep understanding of the connection between miRNAs and lncRNAs during the osteogenic differentiation will strongly improve knowledge into the molecular mechanisms of bone growth and development, ultimately leading to discover innovative diagnostic and therapeutic tools for osteogenic disorders and bone diseases.
Collapse
Affiliation(s)
- Carmen Lanzillotti
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Monica De Mattei
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Chiara Mazziotta
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States
- Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - John Charles Rotondo
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| |
Collapse
|
48
|
Feng L, Yang ZM, Li YC, Wang HX, Lo JHT, Zhang XT, Li G. Linc-ROR promotes mesenchymal stem cells chondrogenesis and cartilage formation via regulating SOX9 expression. Osteoarthritis Cartilage 2021; 29:568-578. [PMID: 33485931 DOI: 10.1016/j.joca.2020.12.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/09/2020] [Accepted: 12/28/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The present study is to characterize the role of long intergenic non-coding RNA, regulator of reprogramming (linc-ROR) in bone marrow mesenchymal stem cell (BMSCs) chondrogenesis, cartilage formation and OA development. METHODS Linc-ROR expression pattern in articular cartilage tissue sample from OA patients were studied by real-time PCR. Linc-ROR lentivirus mediated BMSCs were constructed. In vitro micromass cultured BMSCs chondrogenesis or in vivo MeHA hydrogel encapsulated BMSCs cartilage formation activity were studied. Linc-ROR associating miRNAs which repressed SOX9 expression were characterized by luciferase assay, real-time PCR and Western blot. Linc-ROR was co-transfected with miRNAs into BMSCs to study its rescue effect on SOX9 expression and chondrogenesis activity. RESULTS Linc-ROR was down-regulated in articular cartilage tissue from OA patients and was positively correlated with the expression level of SOX9 (R2 = 0.43). Linc-ROR expression was upregulated during BMSCs chondrogenesis. Linc-ROR ectopic expression significantly promoted in vitro BMSCs chondrogenesis and in vivo cartilage formation activities as revealed by safranin O, alcian blue and COL II staining. The mRNA expression level of chondrogenesis markers including COL II, SOX9 and ACAN were increased, and the hypertrophy markers MMP13 and COL X were decreased upon linc-ROR overexpression in BMSCs. Linc-ROR functioned as a miRNA sponge for miR-138 and miR-145. Both miR-138 and miR-145 suppressed BMSCs chondrogenesis activity and SOX9 expression, while co-expression of linc-ROR displayed a rescuing effect. CONCLUSIONS Taken together, linc-ROR modulated BMSCs chondrogenesis differentiation and cartilage formation by acting as a competing endogenous RNA for miR-138 and miR-145 and activating SOX9 expression. Linc-ROR could be considered as a new diagnostic and therapeutic target for OA treatment.
Collapse
Affiliation(s)
- L Feng
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Z M Yang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Y C Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - H X Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - J H T Lo
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - X T Zhang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - G Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China; MOE Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, SAR, PR China; Department of Orthopaedics and Traumatology, People's Hospital of Baoan District, Shenzhen, PR China.
| |
Collapse
|
49
|
Pan FF, Shao J, Shi CJ, Li ZP, Fu WM, Zhang JF. Apigenin promotes osteogenic differentiation of mesenchymal stem cells and accelerates bone fracture healing via activating Wnt/β-catenin signaling. Am J Physiol Endocrinol Metab 2021; 320:E760-E771. [PMID: 33645251 DOI: 10.1152/ajpendo.00543.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apigenin (API), a natural plant flavone, is abundantly found in common fruits and vegetables. As a bioactive flavonoid, API exhibits several activities including antiproliferation and anti-inflammation. A recent study showed that API could retard osteoporosis progress, indicating its role in the skeletal system. However, the detailed function and mechanism remain obscure. In the present study, API was found to promote osteogenic differentiation of mesenchymal stem cells (MSCs). And further investigation showed that API could enhance the expression of the critical transcription factor β-catenin and several downstream target genes of Wnt signaling, thus activated Wnt/β-catenin signaling. Using a rat femoral fracture model, API was found to improve new bone formation and accelerate fracture healing in vivo. In conclusion, our data demonstrated that API could promote osteogenesis in vitro and facilitate the fracture healing in vivo via activating Wnt/β-catenin signaling, indicating that API may be a promising therapeutic candidate for bone fracture repair.NEW & NOTEWORTHY1) API promoted osteogenic differentiation of human MSCs in vitro; 2) API facilitated bone formation and accelerated fracture healing in vivo; 3) API stimulated Wnt/β-catenin signaling during osteogenesis of human MSCs.
Collapse
Affiliation(s)
- Fei-Fei Pan
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Jiang Shao
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Chuan-Jian Shi
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Zhi-Peng Li
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| | - Wei-Ming Fu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, P.R. China
| | - Jin-Fang Zhang
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, P.R. China
| |
Collapse
|
50
|
LncRNA Mrhl orchestrates differentiation programs in mouse embryonic stem cells through chromatin mediated regulation. Stem Cell Res 2021; 53:102250. [PMID: 33662735 DOI: 10.1016/j.scr.2021.102250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 01/15/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been well-established to act as regulators and mediators of development and cell fate specification programs. LncRNA Mrhl (meiotic recombination hotspot locus) has been shown to act in a negative feedback loop with WNT signaling to regulate male germ cell meiotic commitment. In our current study, we have addressed the role of Mrhl in development and differentiation using mouse embryonic stem cells (mESCs) as our model system of study. Mrhl is a nuclear-localized, chromatin-bound lncRNA with moderately stable expression in mESCs. Transcriptome analyses and loss-of-function phenotype studies revealed dysregulation of developmental processes, lineage-specific transcription factors and key networks along with aberrance in specification of early lineages during differentiation of mESCs. Genome-wide chromatin occupancy studies suggest regulation of chromatin architecture at key target loci through triplex formation. Our studies thus reveal a role for lncRNA Mrhl in regulating differentiation programs in mESCs in the context of appropriate cues through chromatin-mediated responses.
Collapse
|