1
|
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
|
2
|
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
|
3
|
Zhang K, Liu Y, Zhao Z, Shi X, Zhang R, He Y, Zhang H, Wang W. Magnesium-Doped Nano-Hydroxyapatite/Polyvinyl Alcohol/Chitosan Composite Hydrogel: Preparation and Characterization. Int J Nanomedicine 2024; 19:651-671. [PMID: 38269254 PMCID: PMC10807547 DOI: 10.2147/ijn.s434060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024] Open
Abstract
Background Polyvinyl alcohol/Chitosan hydrogel is often employed as a carrier because it is non-toxic, biodegradable, and has a three-dimensional network structure. Meanwhile, Magnesium-doped nano-hydroxyapatite(Mg-nHA) demonstrated high characterization to promote the osteogenic differentiation of bone marrow derived mesenchymal stem cell(BMSCs). Therefore, in order to develop a porous hydrogel scaffold for the application of bone tissue engineering, an appropriate-type Mg-nHA hydrogel scaffold was developed and evaluated. Methods A composite hydrogel containing magnesium-doped nano-hydroxyapatite (Mg-nHA/PVA/CS) was developed using a magnetic stirring-ion exchange method and cyclic freeze-thaw method design, with polyvinyl alcohol and chitosan as the main components. Fourier transform infrared spectra (FTIR), electron energy dispersive spectroscopy (EDS), X-ray photoelectron spectrometer (XPS) and scanning electron microscopy (SEM) were employed to analyze the chemical structure, porosity, and elemental composition of each hydrogels. The equilibrium swelling degree, moisture content, pH change, potential for biomineralization, biocompatibility, the osteogenic potential and magnesium ion release rate of the composite hydrogel were also evaluated. Results SEM analysis revealed a well-defined 3D spatial structure of micropores in the synthesised hydrogel. FTIR analysis showed that doping nanoparticles had little effect on the hydrogel's structure and both the 5% Mg-nHA/PVA/CS and 10% Mg-nHA/PVA/CS groups promoted amide bond formation. EDS observation indicated that the new material exhibited favourable biomineralization ability, with optimal performance seen in the 5% Mg-nHA/PVA/CS group. The composite hydrogel not only displayed favourable water content, enhanced biocompatibility, and porosity (similar to human cancellous bone), but also maintained an equilibrium swelling degree and released magnesium ions that created an alkaline environment around it. Additionally, it facilitated the proliferation of bone marrow mesenchymal stem cells and their osteogenic differentiation. Conclusion The Mg-nHA/PVA/CS hydrogel demonstrates significant potential for application in the field of bone repair, making it an excellent composite material for bone tissue engineering.
Collapse
Affiliation(s)
- Kui Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Yan Liu
- Department of Gynecology, First Affiliated Hospital of Xi ‘an Medical College, Xi’ an, People’s Republic of China
| | - Zhenrui Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Xuewen Shi
- The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Ruihao Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Yixiang He
- The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Huaibin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, People’s Republic of China
| | - Wenji Wang
- Department of Orthopedics, the First Hospital of Lanzhou University, Lanzhou, People’s Republic of China
| |
Collapse
|
4
|
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
|
5
|
Amirlatifi S, Kooshari Z, Salmani K, Fallah Ziyarani M, Azizi S, Ghotbi E, Zolali B. Evaluation of long noncoding RNA (LncRNA) in pathogenesis of HELLP syndrome: diagnostic and future approach. J OBSTET GYNAECOL 2023; 43:2174836. [PMID: 36795605 DOI: 10.1080/01443615.2023.2174836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
HELLP syndrome is a disorder during pregnancy which is defined by elevation of liver enzymes, haemolysis, and low platelet count. This syndrome is a multifactorial one and both genetic and environmental components can have a crucial role in this syndrome's pathogenesis. Long noncoding RNAs (lncRNAs), are defined as long non-protein coding molecules (more than 200 nucleotides), which are functional units in most cellular processes such as cell cycle, differentiation, metabolism and some diseases progression. As these markers discovered, there has been some evidence that they have an important role in the function of some organs, such as placenta; therefore, alteration and dysregulation of these RNAs can develop or alleviate HELLP disorder. Although the role of lncRNAs has been shown in HELLP syndrome, the process is still unclear. In this review, our purpose is to evaluate the association between molecular mechanisms of lncRNAs and HELLP syndrome pathogenicity to elicit some novel approaches for HELLP diagnosis and treatment.
Collapse
Affiliation(s)
- Shahrzad Amirlatifi
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Kooshari
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kiana Salmani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Fallah Ziyarani
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Azizi
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elena Ghotbi
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Bita Zolali
- Clinical Research Development unit (SHACRDU), School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
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
|
7
|
Ai L, Chen L, Tao Y, Wang H, Yi W. Icariin promotes osteogenic differentiation through the mmu_circ_0000349/mmu-miR-138-5p/Jumonji domain-containing protein-3 axis. Heliyon 2023; 9:e21885. [PMID: 38045146 PMCID: PMC10692785 DOI: 10.1016/j.heliyon.2023.e21885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/22/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Circular RNAs (circRNAs) regulate Jumonji domain-containing protein-3 (JMJD3) by sponging with microRNAs (miRNAs). This study aimed to investigate the role of icariin on specific circRNA/miRNA/JMJD3 axis in osteogenic differentiation of MC3T3-E1 cells. CircRNA sequencing was performed on the MC3T3-E1 cells induced by osteogenic differentiation medium for 1 d (negative control (NC) group) and 14 d (osteogenesis group). And mmu_circ_0000349 was verified using Sanger sequencing, ribonuclease R degradation, and actinomycin D assay. The function of mmu_circ_0000349 was validated by detecting the expressions of osteogenic differentiation markers, alkaline phosphatase (ALP), and runt-related transcription (RUNX2), via real-time quantitative PCR (qPCR) and Western blotting or ALP and alizarin red staining assay. Dual luciferase reporter gene assay confirmed the relationship between mmu_circ_0000349 and mmu-miR-138-5p (or mmu-miR-138-5p and JMJD3). Meanwhile, the JMJD3 binding to mmu_circ_0000349 was screened using an RNA pull-down assay. qPCR and Western blotting confirmed the effect of icariin on the mmu_circ_0000349/mmu-miR-138-5p/JMJD3 axis and osteogenic differentiation. As MC3T3-E1 osteogenic differentiation progressed, the JMJD3 expression level increased. A total of 361 circRNAs exhibited differences between the NC and osteogenesis groups. After validation, mmu_circ_0000349 was further analyzed as it exhibited the largest expression. And mmu_circ_0000349 was identified as a stable circular structure. Overexpression of mmu_circ_0000349 increased the expression levels of ALP and RUNX2, enhanced ALP activity, and increased the number of mineralized nodules; contrarily, inhibition of mmu_circ_0000349 exerted opposite effects. The data also confirmed that mmu_circ_0000349 regulated JMJD3 by sponging with mmu-miR-138-5p. With the increase in icariin concentration and time for treatment, the expression levels of mmu_circ_0000349, JMJD3, ALP, and RUNX2 also increased, whereas that of mmu-miR-138-5p decreased. In conclusion, Icariin promoted osteogenic differentiation by regulating the mmu_circ_0000349/mmu-miR-138-5p/JMJD3 pathway. Therefore, this provides a theoretical basis for the treatment of diseases related to osteogenic differentiation.
Collapse
Affiliation(s)
- Liang Ai
- Department of TCM, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Liudan Chen
- Department of TCM and Acupuncture, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yangu Tao
- Department of TCM, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Haibin Wang
- Department of Orthopaedics, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Weimin Yi
- Department of TCM and Acupuncture, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| |
Collapse
|
8
|
Zhang Y, Chen X, Yang X, Huang L, Qiu X. Mesenchymal Stem Cell-Derived from Dental Tissues-Related lncRNAs: A New Regulator in Osteogenic Differentiation. J Tissue Eng Regen Med 2023; 2023:4622584. [PMID: 40226409 PMCID: PMC11919082 DOI: 10.1155/2023/4622584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/12/2023] [Accepted: 06/22/2023] [Indexed: 04/15/2025]
Abstract
Odontogenic stem cells are mesenchymal stem cells (MSCs) with multipotential differentiation potential from different dental tissues. Their osteogenic differentiation is of great significance in bone tissue engineering. In recent years, it has been found that long noncoding RNAs (lncRNAs) participate in regulating the osteoblastic differentiation of stem cells at the epigenetic level, transcriptional level, and posttranscriptional level. We reviewed the existing lncRNA related to the osteogenic differentiation of odontogenic stem cells and emphasized the critical mechanism of lncRNA in the osteogenic differentiation of odontogenic stem cells. These findings are expected to be an important target for promoting osteoblastic differentiation of odontogenic stem cells in bone regeneration therapy with lncRNA.
Collapse
Affiliation(s)
- Yinchun Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - Xuan Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - XiaoXia Yang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - Lei Huang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| | - Xiaoling Qiu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangdong 510280, China
| |
Collapse
|
9
|
Fu Y, Zhang JB, Han DX, Wang HQ, Liu JB, Xiao Y, Jiang H, Gao Y, Yuan B. CiRS-187 regulates BMPR2 expression by targeting miR-187 in bovine cumulus cells treated with BMP15 and GDF9. Theriogenology 2023; 197:62-70. [PMID: 36470111 DOI: 10.1016/j.theriogenology.2022.10.034] [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: 03/10/2022] [Revised: 06/13/2022] [Accepted: 10/28/2022] [Indexed: 11/23/2022]
Abstract
Circular RNAs (circRNAs) play vital roles in regulating biological processes. However, the contributions of circRNAs to BMPR2 regulation during follicle development remain unknown. In this study, we first verified the optimal conditions for BMP15 and GDF9 treatment in bovine cumulus cells. Then, we screened and identified candidate microRNAs (miRNAs) that may target the BMPR2 3'UTR with TargetScan, a luciferase reporter assay and RT-qPCR. Next, we transfected miR-187 into bovine cumulus cells, and the results showed that miR-187 regulated BMPR2 and inhibited its expression. To explore the competing endogenous RNA (ceRNA) mechanism, we predicted the sponging circRNAs of miR-187 and identified ciRS-187. We further detected miR-187 and BMPR2 expression and apoptosis levels upon knockdown of ciRS-187 and found that ciRS-187 upregulated BMPR2 expression. The results provide a theoretical basis for a ceRNA mechanism of circRNAs related to follicle development.
Collapse
Affiliation(s)
- Yao Fu
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China; National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jia-Bao Zhang
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Dong-Xu Han
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Hao-Qi Wang
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Jian-Bo Liu
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Yue Xiao
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Hao Jiang
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Yan Gao
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China
| | - Bao Yuan
- Department of Laboratory Animal Science, College of Animal Sciences, Jilin University, Changchun, 130062, China.
| |
Collapse
|
10
|
Zhang Q, Liu Y, Li J, Wang J, Liu C. Recapitulation of growth factor-enriched microenvironment via BMP receptor activating hydrogel. Bioact Mater 2023; 20:638-650. [PMID: 35846838 PMCID: PMC9270210 DOI: 10.1016/j.bioactmat.2022.06.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/31/2022] [Accepted: 06/15/2022] [Indexed: 11/18/2022] Open
Abstract
Exposure to a growth factor abundant milieu has remarkable regenerative and rejuvenating effects on organ diseases, tissue damage, and regeneration, including skeletal system defects and bone regeneration. Although the introduction of candidate growth factors into relevant fields has been reported, their regenerative effects remain unsatisfactory, mainly because of the experimental challenges with limited types of growth factors, elusive dosage adjustment, and asynchronous stem cell activation with cytokine secretion. Here, an innovative hydrogel recapitulating a growth factor-enriched microenvironment (GEM) for regenerative advantage, is reported. This sulfated hydrogel includes bone morphogenetic protein-2 (BMP-2), an essential growth factor in osteogenesis, to direct mesenchymal stem cell (MSC) differentiation, stimulate cell proliferation, and improve bone formation. The semi-synthetic hydrogel, sulfonated gelatin (S-Gelatin), can amplify BMP-2 signaling in mouse MSCs by enhancing the binding between BMP-2 and BMP-2 type II receptors (BMPR2), which are located on MSC nuclei and activated by the hydrogel. Importantly, the dramatically improved cytokine secretion of MSCs throughout regeneration confirms the growth factor-acquiring potential of S-Gelatin/rhBMP-2 hydrogel, leading to the vascularization enhancement. These findings provide a new strategy to achieve an in situ GEM and accelerated bone regeneration by amplifying the regenerative capacity of rhBMP-2 and capturing endogenous growth factors.
Collapse
Affiliation(s)
- Qinghao Zhang
- Material Science and Engineering School, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Meilong Road 130, Shanghai, 200237, PR China
| | - Yuanda Liu
- Material Science and Engineering School, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Meilong Road 130, Shanghai, 200237, PR China
| | - Jie Li
- Material Science and Engineering School, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Meilong Road 130, Shanghai, 200237, PR China
| | - Jing Wang
- Material Science and Engineering School, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Material Science and Engineering School, East China University of Science and Technology, Shanghai, 200237, PR China
- Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai, 200237, PR China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Meilong Road 130, Shanghai, 200237, PR China
| |
Collapse
|
11
|
Breulmann FL, Hatt LP, Schmitz B, Wehrle E, Richards RG, Della Bella E, Stoddart MJ. Prognostic and therapeutic potential of microRNAs for fracture healing processes and non-union fractures: A systematic review. Clin Transl Med 2023; 13:e1161. [PMID: 36629031 PMCID: PMC9832434 DOI: 10.1002/ctm2.1161] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Approximately 10% of all bone fractures result in delayed fracture healing or non-union; thus, the identification of biomarkers and prognostic factors is of great clinical interest. MicroRNAs (miRNAs) are known to be involved in the regulation of the bone healing process and may serve as functional markers for fracture healing. AIMS AND METHODS This systematic review aimed to identify common miRNAs involved in fracture healing or non-union fractures using a qualitative approach. A systematic literature search was performed with the keywords 'miRNA and fracture healing' and 'miRNA and non-union fracture'. Any original article investigating miRNAs in fracture healing or non-union fractures was screened. Eventually, 82 studies were included in the qualitative analysis for 'miRNA and fracture healing', while 19 were selected for the 'miRNA and fracture non-union' category. RESULTS AND CONCLUSIONS Out of 151 miRNAs, miR-21, miR-140 and miR-214 were the most investigated miRNAs in fracture healing in general. miR-31-5p, miR-221 and miR-451-5p were identified to be regulated specifically in non-union fractures. Large heterogeneity was detected between studies investigating the role of miRNAs in fracture healing or non-union in terms of patient population, sample types and models used. Nonetheless, our approach identified some miRNAs with the potential to serve as biomarkers for non-union fractures, including miR-31-5p, miR-221 and miR-451-5p. We provide a discussion of involved pathways and suggest on alignment of future research in the field.
Collapse
Affiliation(s)
- Franziska Lioba Breulmann
- AO Research Institute DavosDavos PlatzSwitzerland
- Department of Orthopedic Sports MedicineKlinikum Rechts der IsarTechnical University of MunichMunichGermany
| | - Luan Phelipe Hatt
- AO Research Institute DavosDavos PlatzSwitzerland
- Institute for BiomechanicsETH ZürichZurichSwitzerland
| | - Boris Schmitz
- Department of Rehabilitation SciencesFaculty of HealthUniversity of Witten/HerdeckeWittenGermany
- DRV Clinic KönigsfeldCenter for Medical RehabilitationEnnepetalGermany
| | - Esther Wehrle
- AO Research Institute DavosDavos PlatzSwitzerland
- Institute for BiomechanicsETH ZürichZurichSwitzerland
| | - Robert Geoff Richards
- AO Research Institute DavosDavos PlatzSwitzerland
- Faculty of MedicineMedical Center‐Albert‐Ludwigs‐University of FreiburgAlbert‐Ludwigs‐University of FreiburgFreiburgGermany
| | | | - Martin James Stoddart
- AO Research Institute DavosDavos PlatzSwitzerland
- Faculty of MedicineMedical Center‐Albert‐Ludwigs‐University of FreiburgAlbert‐Ludwigs‐University of FreiburgFreiburgGermany
| |
Collapse
|
12
|
Zhang Q, Long Y, Jin L, Li C, Long J. Non-coding RNAs regulate the BMP/Smad pathway during osteogenic differentiation of stem cells. Acta Histochem 2023; 125:151998. [PMID: 36630753 DOI: 10.1016/j.acthis.2023.151998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
MicroRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) are involved in the regulation of bone metabolism. The BMP/Smad pathway is a key signaling pathway for classical regulation of osteogenic differentiation. Non-coding RNAs (ncRNAs) and the BMP/Smad pathway both have important roles for osteogenic differentiation of stem cells, bone regeneration, and development of bone diseases. There is increasing evidence that ncRNAs interact with the BMP/Smad pathway to regulate not only osteogenic differentiation of stem cells but also progression of bone diseases, such as osteoporosis (OP), myeloma, and osteonecrosis of the femoral head (ONFH), by controlling the expression of bone disease-related genes. Therefore, ncRNAs that interact with BMP/Smad pathway molecules are potential targets for bone regeneration as well as bone disease diagnosis, prevention, and treatment. However, despite extensive studies on ncRNAs associated with the BMP/Smad pathway and osteogenic differentiation of stem cells, there is a lack of comparability. Moreover, some bone disease-associated ncRNAs with low abundance can be difficult to detect and there is a lack of mature delivery systems for their stable translocation to target sites, thus limiting their application. In this review, we summarize the research progress on interactions between ncRNAs and the BMP/Smad pathway during osteogenic differentiation of various stem cells and in the regulation of bone regeneration and bone diseases.
Collapse
Affiliation(s)
- Qiuling Zhang
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Yifei Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China
| | - Liangyu Jin
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Chenghao Li
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Jie Long
- The State Key Laboratory of Oral Diseases, Sichuan University, Chengdu 610041, PR China; Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
13
|
Wang Z, Zhang J, Hu J, Yang G. Gene-activated titanium implants for gene delivery to enhance osseointegration. BIOMATERIALS ADVANCES 2022; 143:213176. [PMID: 36327825 DOI: 10.1016/j.bioadv.2022.213176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/22/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Osseointegration is the direct and intimate contact between mineralized tissue and titanium implant at the bone-implant interface. Early establishment and stable maintenance of osseointegration is the key to long-term implant success. However, in patients with compromised conditions such as osteoporosis and patients beginning early load-bearing activities such as walking, lower osseointegration around titanium implants is often observed, which might result in implant early failure. Gene-activated implants show an exciting prospect of combining gene delivery and biomedical implants to solve the problems of poor osseointegration formation, overcoming the shortcomings of protein therapy, including rapid degradation and overdose adverse effects. The conception of gene-activated titanium implants is based on "gene-activated matrix" (GAM), which means scaffolds using non-viral vectors for in situ gene delivery to achieve a long-term and efficient transfection of target cells. Current preclinical studies in animal models have shown that plasmid DNA (pDNA), microRNA (miRNA), and small interference RNA (siRNA) functionalized titanium implants can enhance osseointegration with safety and efficiency, leading to the expectation of applying this technique in dental and orthopedic clinical scenarios. This review aims to comprehensively summarize fabrication strategies, current applications, and futural outlooks of gene-activated implants, emphasizing nucleic acid targets, non-viral vectors, implant surface modification techniques, nucleic acid/vector complexes loading strategies.
Collapse
Affiliation(s)
- Zhikang Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jinxing Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
14
|
Ranjbarnejad F, Khazaei M, Shahryari A, Khazaei F, Rezakhani L. Recent advances in gene therapy for bone tissue engineering. J Tissue Eng Regen Med 2022; 16:1121-1137. [PMID: 36382408 DOI: 10.1002/term.3363] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 10/05/2022] [Accepted: 10/23/2022] [Indexed: 11/18/2022]
Abstract
Autografting, a major treatment for bone fractures, has potential risks related to the required surgery and disease transmission. Bone morphogenetic proteins (BMPs) are the most common osteogenic factors used for bone-healing applications. However, BMP delivery can have shortcomings such as a short half-life and the high cost of manufacturing the recombinant proteins. Gene delivery methods have demonstrated promising alternative strategies for producing BMPs or other osteogenic factors using engineered cells. These approaches can also enable temporal overexpression and local production of the therapeutic genes in the target tissues. This review addresses recent progress on engineered viral, non-viral, and RNA-mediated gene delivery systems that are being used for bone repair and regeneration. Advances in clustered regularly interspaced short palindromic repeats/Cas9 genome engineering for bone tissue regeneration also is discussed.
Collapse
Affiliation(s)
- Fatemeh Ranjbarnejad
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Alireza Shahryari
- Tools for Bio-Imaging, Max-Planck-Institute for Biological Intelligence, Martinsried, Germany
| | - Fatemeh Khazaei
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Li D, Zhang D, Yuan Q, Liu L, Li H, Xiong L, Guo X, Yan Y, Yu K, Dai Y, Xiao T, Li Y, Wen C. In vitro and in vivo assessment of the effect of biodegradable magnesium alloys on osteogenesis. Acta Biomater 2022; 141:454-465. [PMID: 34971787 DOI: 10.1016/j.actbio.2021.12.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/09/2021] [Accepted: 12/24/2021] [Indexed: 12/17/2022]
Abstract
Magnesium (Mg) and some of its alloys are considered promising biodegradable metallic biomaterials for bone implant applications. The osteogenesis effect of Mg alloys is widely reported; however, the underlying mechanisms are still not clear. In this study, pure Mg, Mg-3Zn, and Mg-2Zn-1Mn were prepared, and their degradation behavior, biocompatibility, and osteogenesis effect were systematically assessed both in vitro and in vivo. Primary rat bone marrow-derived mesenchymal stem cells (BMSCs) were used to evaluate the biocompatibility of the prepared Mg alloys, and a rat femur fracture model was used to assess the stimulating effect of these alloys on bone-tissue formation. Mg-2Zn-1Mn showed higher corrosion resistance and more stable degradation behavior than pure Mg and Mg-3Zn. Extracts of the three materials showed significant stimulating effects on osteogenic differentiation of BMSCs along with non-cytotoxicity. Implantation of Mg-2Zn-1Mn wires into the femur of rats demonstrated superior histocompatibility, stable degradation, and notable promotion of osteogenesis without systemic toxicity. Moreover, the results of both in vitro and in vivo assessments demonstrated that bone morphogenetic proteins and fibroblast growth factor receptors are involved in the stimulating effect of Mg alloys. STATEMENT OF SIGNIFICANCE: This work reports the degradation behavior, biocompatibility, and osteogenic effect of pure Mg and Mg-3Zn and Mg-2Zn-1Mn alloys in both in vitro and in vivo conditions. Mg-2Zn-1Mn showed higher corrosion resistance and more stable degradation behavior than pure Mg and Mg-3Zn. The extracts of the three materials showed a significant stimulating effect on osteogenic differentiation of rat bone marrow-derived mesenchymal stem cells (BMSCs) along with non-cytotoxicity. Mg-2Zn-1Mn wires implanted into the femur of rats showed good histocompatibility, stable degradation, and notable promotion of osteogenesis without systemic toxicity. The results of the present study suggest that bone morphogenetic proteins (BMPs) and fibroblast growth factor receptors (FGFRs) are involved in the stimulating effect of Mg alloys on osteogenesis.
Collapse
Affiliation(s)
- Ding Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 410011, China; Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha 410011, China
| | - Dechuang Zhang
- School of Materials Science and Engineering, Xiangtan University, Xiangtan, Hunan 411105, China.
| | - Qi Yuan
- Department of Hepatopathy, The Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410002, China
| | - Lihong Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 410011, China; Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha 410011, China
| | - Hui Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 410011, China; Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha 410011, China
| | - Liang Xiong
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 410011, China; Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha 410011, China
| | - Xiaoning Guo
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 410011, China; Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha 410011, China
| | - Yang Yan
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Kun Yu
- School of Materials Science and Engineering, Central South University, Changsha 410083, China
| | - Yilong Dai
- School of Materials Science and Engineering, Central South University, Changsha 410083, China.
| | - Tao Xiao
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 410011, China; Orthopedic Biomedical Materials Engineering Laboratory of Hunan Province, Changsha 410011, China.
| | - Yuncang Li
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia
| | - Cuie Wen
- School of Engineering, RMIT University, Melbourne, Victoria 3001, Australia.
| |
Collapse
|
16
|
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
|
17
|
Sun Y, Helmholz H, Will O, Damm T, Wiese B, Luczak M, Peschke E, Luthringer-Feyerabend B, Ebel T, Hövener JB, Glüer C, Willumeit-Römer R. Dynamic in vivo monitoring of fracture healing process in response to magnesium implant with multimodal imaging: Pilot longitudinal study in a rat external fixation model. Biomater Sci 2022; 10:1532-1543. [DOI: 10.1039/d2bm00051b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Rodent models are commonly used in pre-clinical research of magnesium (Mg) -based and other types of biomaterials for fracture treatment. Most studies selected unstable fixation methods, and there is a...
Collapse
|
18
|
Yang W, Zhu W, Yang Y, Guo M, Qian H, Jiang W, Chen Y, Lian C, Xu Z, Bai H, Chen T, Zhang J. Exosomal miR-100-5p inhibits osteogenesis of hBMSCs and angiogenesis of HUVECs by suppressing the BMPR2/Smad1/5/9 signalling pathway. Stem Cell Res Ther 2021; 12:390. [PMID: 34256859 PMCID: PMC8278698 DOI: 10.1186/s13287-021-02438-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 06/09/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Nontraumatic osteonecrosis of the femoral head (NONFH) is a common, progressive, and refractory orthopaedic disease. Decreased osteogenesis and angiogenesis are considered the main factors in the pathogenesis of NONFH. We aimed to figure out whether exosomes and exosomal miRNA from necrotic bone tissues of patients with NONFH are involved in the pathogenesis of NONFH and reveal the underlying mechanisms. METHODS RT-PCR and western blotting (WB) were used to detect the expression of osteogenic, adipogenic, and angiogenic markers. ALP staining and Alizarin Red S (ARS) staining were used to evaluate osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs). Oil Red O staining was performed to assess the adipocyte deposition. A tube formation assay was used to study angiogenesis of human umbilical vascular endothelial cells (HUVECs). H&E staining and immunohistochemistry (IHC) staining were used to detect the effect of the NONFH exosomes in vivo. MicroRNA sequencing was conducted to identify potential regulators in the NONFH exosomes. The target relationship between miR-100-5p and BMPR2 was predicted and confirmed by a dual luciferase reporter assay and WB. RESULTS The NONFH exosomes reduced the osteogenic differentiation of hBMSCs and angiogenesis of HUVECs. In addition, the injection of the NONFH exosomes caused thinning and disruption of bone trabeculae in the femoral heads of rats. MiR-100-5p expression was upregulated in the NONFH exosomes and inhibited the osteogenesis of hBMSCs and angiogenesis of HUVECs by targeting BMPR2 and suppressing the BMPR2/SMAD1/5/9 signalling pathway. Silencing miR-100-5p expression rescued the reduction in osteogenesis and angiogenesis caused by the NONFH exosomes by activating the BMPR2/SMAD1/5/9 signalling pathway. CONCLUSION The NONFH exosomal miR-100-5p can lead to NONFH-like damage by targeting BMPR2 and suppressing the BMPR2/SMAD1/5/9 signalling pathway, which may be involved in the pathophysiological mechanisms of nontraumatic osteonecrosis of the femoral head (NONFH).
Collapse
Affiliation(s)
- Wu Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Weiwen Zhu
- Department of Orthopedics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yunfei Yang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Minkang Guo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Husun Qian
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Weiqian Jiang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yu Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengjie Lian
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zijie Xu
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Haobo Bai
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Tingmei Chen
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, 400016, China
| | - Jian Zhang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
19
|
Damiati LA, El-Messeiry S. An Overview of RNA-Based Scaffolds for Osteogenesis. Front Mol Biosci 2021; 8:682581. [PMID: 34169095 PMCID: PMC8217814 DOI: 10.3389/fmolb.2021.682581] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/06/2021] [Indexed: 12/20/2022] Open
Abstract
Tissue engineering provides new hope for the combination of cells, scaffolds, and bifactors for bone osteogenesis. This is achieved by mimicking the bone's natural behavior in recruiting the cell's molecular machinery for our use. Many researchers have focused on developing an ideal scaffold with specific features, such as good cellular adhesion, cell proliferation, differentiation, host integration, and load bearing. Various types of coating materials (organic and non-organic) have been used to enhance bone osteogenesis. In the last few years, RNA-mediated gene therapy has captured attention as a new tool for bone regeneration. In this review, we discuss the use of RNA molecules in coating and delivery, including messenger RNA (mRNA), RNA interference (RNAi), and long non-coding RNA (lncRNA) on different types of scaffolds (such as polymers, ceramics, and metals) in osteogenesis research. In addition, the effect of using gene-editing tools-particularly CRISPR systems-to guide RNA scaffolds in bone regeneration is also discussed. Given existing knowledge about various RNAs coating/expression may help to understand the process of bone formation on the scaffolds during osseointegration.
Collapse
Affiliation(s)
- Laila A. Damiati
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Sarah El-Messeiry
- Department of Genetics, Faculty of Agriculture, Alexandria University, Alexandria, Egypt
| |
Collapse
|
20
|
Guo G, Gou Y, Jiang X, Wang S, Wang R, Liang C, Yang G, Wang T, Yu A, Zhu G. Long Non-coding RNAs in Traumatic Brain Injury Accelerated Fracture Healing. Front Surg 2021; 8:663377. [PMID: 34150839 PMCID: PMC8211774 DOI: 10.3389/fsurg.2021.663377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/28/2021] [Indexed: 11/13/2022] Open
Abstract
It is commonly observed that patients with bone fracture concomitant with traumatic brain injury (TBI) had significantly increased fracture healing, but the underlying mechanisms were not fully revealed. Long non-coding RNAs (lncRNAs) are known to play complicated roles in bone homeostasis, but their role in TBI accelerated fracture was rarely reported. The present study was designed to determine the role of lncRNAs in TBI accelerated fracture via transcriptome sequencing and further bioinformatics analyses. Blood samples from three fracture-only patients, three fracture concomitant with TBI patients, and three healthy controls were harvested and were subsequently subjected to transcriptome lncRNA sequencing. Differentially expressed genes were identified, and pathway enrichment was performed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. High-dimensional data visualization by self-organizing map (SOM) machine learning was applied to further interpret the data. An xCell method was then used to predict cellular behavior in all samples based on gene expression profiles, and an lncRNA-cell interaction network was generated. A total of 874 differentially expressed genes were identified, of which about 26% were lncRNAs. Those identified lncRNAs were mainly enriched on TBI-related and damage repair-related pathways. SOM analyses revealed that those differentially expressed lncRNAs could be divided into three major module implications and were mainly enriched on transcriptional regulation and immune-related signal pathways, which promote us to further explore cellular behaviors based on differentially expressed lncRNAs. We have predicted that basophils, CD8+ T effector memory cells, B cells, and naïve B cells were significantly downregulated, while microvascular endothelial cells were predicted to be significantly upregulated in the Fr/TBI group, was the lowest and highest, respectively. ENSG00000278905, ENSG00000240980, ENSG00000255670, and ENSG00000196634 were the most differentially expressed lncRNAs related to all changes of cellular behavior. The present study has revealed for the first time that several critical lncRNAs may participate in TBI accelerated fracture potentially via regulating cellular behaviors of basophils, cytotoxic T cells, B cells, and endothelial cells.
Collapse
Affiliation(s)
- Guoning Guo
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Yajun Gou
- Department of Orthopedic, Shapingba District Hospital, Chongqing, China
| | - Xingyu Jiang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Shuhong Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Ruilie Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Changqiang Liang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Guang Yang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Tinggang Wang
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Guizhou, China
| | - Guoyan Zhu
- Department of Health Management, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
21
|
Guo B, Zhu X, Li X, Yuan CF. The Roles of LncRNAs in Osteogenesis, Adipogenesis and Osteoporosis. Curr Pharm Des 2021; 27:91-104. [PMID: 32634074 DOI: 10.2174/1381612826666200707130246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/28/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Osteoporosis (OP) is the most common bone disease, which is listed by the World Health Organization (WHO) as the third major threat to life and health among the elderly. The etiology of OP is multifactorial, and its potential regulatory mechanism remains unclear. Long non-coding RNAs (LncRNAs) are the non-coding RNAs that are over 200 bases in the chain length. Increasing evidence indicates that LncRNAs are the important regulators of osteogenic and adipogenic differentiation, and the occurrence of OP is greatly related to the dysregulation of the bone marrow mesenchymal stem cells (BMSCs) differentiation lineage. Meanwhile, LncRNAs affect the occurrence and development of OP by regulating OP-related biological processes. METHODS In the review, we summarized and analyzed the latest findings of LncRNAs in the pathogenesis, diagnosis and related biological processes of OP. Relevant studies published in the last five years were retrieved and selected from the PubMed database using the keywords of LncRNA and OP. RESULTS/CONCLUSION The present study aimed to examine the underlying mechanisms and biological roles of LncRNAs in OP, as well as osteogenic and adipogenic differentiation. Our results contributed to providing new clues for the epigenetic regulation of OP, making LncRNAs the new targets for OP therapy.
Collapse
Affiliation(s)
- Bo Guo
- China Three Gorges University, RenHe Hospital, Yichang, China
| | - Xiaokang Zhu
- China Three Gorges University, RenHe Hospital, Yichang, China
| | - Xinzhi Li
- China Three Gorges University, RenHe Hospital, Yichang, China
| | - C F Yuan
- Department of Biochemistry, China Three Gorges University, Yichang, China
| |
Collapse
|
22
|
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
|
23
|
Komatsu DE, Duque E, Hadjiargyrou M. MicroRNAs and fracture healing: Pre-clinical studies. Bone 2021; 143:115758. [PMID: 33212318 PMCID: PMC7769985 DOI: 10.1016/j.bone.2020.115758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/28/2022]
Abstract
During the past several years, pre-clinical experiments have established that microRNAs (miRNAs), small non-coding RNAs, serve as key regulatory molecules of fracture healing. Their easy modulation with agonists and antagonists make them highly desirable targets for future therapeutic strategies, especially for pathophysiologic fractures that either do not heal (nonunions) or are delayed. It is now well documented that these problematic fractures lead to human suffering and impairment of life quality. Additionally, financial difficulties are also encountered as work productivity decreases and income is reduced. Moreover, targeting miRNAs may also be an avenue to enhancing normal physiological fracture healing. Herein we present the most current knowledge of the involvement of miRNAs during fracture healing in pre-clinical studies. Following a brief description on the nature of miRNAs and of the fracture healing process, we present data from studies focusing specifically, on miRNA regulation of osteoblast differentiation and osteogenesis (within the context of known signaling pathways), chondrocytes, angiogenesis, and apoptosis, all critical to successful bone repair. Further, we also discuss miRNAs and exosomes. We hope that this manuscript serves as a comprehensive review that will facilitate basic/translational scientists in the orthopaedic arena to realize and further decipher the biological and future therapeutic impact of these small regulatory RNA molecules, especially as they relate to the molecular events of each of the major phases of fracture healing.
Collapse
Affiliation(s)
- David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Edie Duque
- Department of Orthopaedics and Rehabilitation, Stony Brook University, United States of America
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, United States of America.
| |
Collapse
|
24
|
Wang J, Liu S, Shi J, Liu H, Li J, Zhao S, Yi Z. The Role of lncRNAs in Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells. Curr Stem Cell Res Ther 2020; 15:243-249. [PMID: 31880266 DOI: 10.2174/1574888x15666191227113742] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 02/01/2023]
Abstract
Bone Marrow Mesenchymal Stem Cells (BMSCs) are one of the primary cells found in the bone marrow, and they can differentiate into osteoblasts, chondrocytes, adipocytes and even myoblasts, and are, therefore, considered pluripotent cells. Because of their multipotential differentiation, selfrenewal capability, immunomodulation and other potential activities, BMSCs have become an important source of seed cells for gene therapy, tissue engineering, cell replacement therapy and regenerative medicine. Long non-coding RNA (lncRNA) is an RNA molecule greater than 200 nucleotides in length that is expressed in a variety of species, including animals, plants, yeast, prokaryotes, and viruses, but lacks an apparent open reading frame, and does not have the function of translation into proteins. Many studies have shown that lncRNAs play an important role in the osteogenic differentiation of BMSCs. Here, we describe the role of lncRNAs in the osteogenic differentiation of BMSCs, in order to provide a new theoretical and experimental basis for bone tissue engineering and clinical treatment.
Collapse
Affiliation(s)
- Jicheng Wang
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China.,Xi'an Medical University, Xi'an 710068, China
| | - Shizhang Liu
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Jiyuan Shi
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Huitong Liu
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Jingyuan Li
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Song Zhao
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China.,Xi'an Medical University, Xi'an 710068, China
| | - Zhi Yi
- Department of Orthopaedic, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| |
Collapse
|
25
|
Jiang H, Jia P. MiR-153-3p inhibits osteogenic differentiation of periodontal ligament stem cells through KDM6A-induced demethylation of H3K27me3. J Periodontal Res 2020; 56:379-387. [PMID: 33368310 DOI: 10.1111/jre.12830] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/23/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND OBJECTIVE Periodontal ligament stem cells (PDLSCs) have potential for osteogenic differentiation and show a great foreground in treating bone diseases. Histone three lysine 27 (H3K27) demethylase lysine demethylase 6A (KDM6A) is a critical epigenetic modifier and plays an important role in regulating osteogenic differentiation. Multiple microRNAs have been found to play important roles in osteogenesis. The aim of this study was to explore the mechanisms underlying the roles of miR-153-3p and KDM6A in PDLSC osteogenesis. METHODS The levels of the osteogenic markers alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and osteopontin (OPN) were measured by western blotting. Osteoblast activity and mineral deposition were detected by ALP and Alizarin red S (ARS) staining. The levels of miR-153-3p and KDM6A were measured by quantitative real-time PCR (qRT-PCR). A luciferase reporter assay was used to confirm the interaction between KDM6A and miR-153-3p. Gain-of-function and loss-of-function assays were performed to identify the roles of miR-153-3p and KDM6A in the osteogenic differentiation of PDLSCs. RESULTS In osteogenic PDLSCs, the expression of KDM6A, ALP, Runx2, and OPN was upregulated, whereas that of miR-153-3p was downregulated. miR-153-3p downregulation or KDM6A overexpression promoted the osteogenic differentiation of PDLSCs, as demonstrated by increases in ALP activity, matrix mineralization, and ALP, Runx2, and OPN expression. KDM6A was confirmed to be a target of miR-153-3p, and KDM6A overexpression reversed the inhibitory effect of miR-153-3p mimic on PDLSC osteogenesis. KDM6A promoted ALP, Runx2, and OPN expression through the demethylation of H3K27me3 on the promoter regions of these genes. CONCLUSION miR-153-3p inhibited PDLSC osteogenesis by targeting KDM6A and inhibiting ALP, Runx2, and OPN transcription. These findings provide latent hope for PDLSCs application in periodontal therapy.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Stomatology, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Peizeng Jia
- Department of Orthodontics, Peking University School of Stomatology, Beijing, China
| |
Collapse
|
26
|
Zhang J, Shang Z, Jiang Y, Zhang K, Li X, Ma M, Li Y, Ma B. Biodegradable metals for bone fracture repair in animal models: a systematic review. Regen Biomater 2020; 8:rbaa047. [PMID: 33732493 PMCID: PMC7947587 DOI: 10.1093/rb/rbaa047] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/27/2020] [Accepted: 09/16/2020] [Indexed: 12/19/2022] Open
Abstract
Biodegradable metals hold promises for bone fracture repair. Their clinical translation requires pre-clinical evaluations including animal studies, which demonstrate the safety and performance of such materials prior to clinical trials. This evidence-based study investigates and analyzes the performance of bone fractures repair as well as degradation properties of biodegradable metals in animal models. Data were carefully collected after identification of population, interventions, comparisons, outcomes and study design, as well as inclusion criteria combining biodegradable metals and animal study. Twelve publications on pure Mg, Mg alloys and Zn alloys were finally included and reviewed after extraction from a collected database of 2122 publications. Compared to controls of traditional non-degradable metals or resorbable polymers, biodegradable metals showed mixed or contradictory outcomes of fracture repair and degradation in animal models. Although quantitative meta-analysis cannot be conducted because of the data heterogeneity, this systematic review revealed that the quality of evidence for biodegradable metals to repair bone fractures in animal models is 'very low'. Recommendations to standardize the animal studies of biodegradable metals were proposed. Evidence-based biomaterials research could help to both identify reliable scientific evidence and ensure future clinical translation of biodegradable metals for bone fracture repair.
Collapse
Affiliation(s)
- Jiazhen Zhang
- State Key Laboratory of Nonferrous Metals and Process, GRINM Group Corporation Limited (GRINM), No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China.,GRIMAT Engineering Institute Co., Ltd, No. 11, Xingke East St., Yanqi Economic Development Zone, Huairou District, Beijing 101407, P.R. China.,General Research Institute for Nonferrous Metals, No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China
| | - Zhizhong Shang
- School of Basic Medical Sciences, Evidence-Based Medicine Center, Lanzhou University, No 199, Donggang West Road, Chengguan District, Lanzhou 730000, P. R. China
| | - Yanbiao Jiang
- School of Basic Medical Sciences, Evidence-Based Medicine Center, Lanzhou University, No 199, Donggang West Road, Chengguan District, Lanzhou 730000, P. R. China
| | - Kui Zhang
- State Key Laboratory of Nonferrous Metals and Process, GRINM Group Corporation Limited (GRINM), No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China.,GRIMAT Engineering Institute Co., Ltd, No. 11, Xingke East St., Yanqi Economic Development Zone, Huairou District, Beijing 101407, P.R. China.,General Research Institute for Nonferrous Metals, No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China
| | - Xinggang Li
- State Key Laboratory of Nonferrous Metals and Process, GRINM Group Corporation Limited (GRINM), No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China.,GRIMAT Engineering Institute Co., Ltd, No. 11, Xingke East St., Yanqi Economic Development Zone, Huairou District, Beijing 101407, P.R. China.,General Research Institute for Nonferrous Metals, No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China
| | - Minglong Ma
- State Key Laboratory of Nonferrous Metals and Process, GRINM Group Corporation Limited (GRINM), No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China.,GRIMAT Engineering Institute Co., Ltd, No. 11, Xingke East St., Yanqi Economic Development Zone, Huairou District, Beijing 101407, P.R. China.,General Research Institute for Nonferrous Metals, No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China
| | - Yongjun Li
- State Key Laboratory of Nonferrous Metals and Process, GRINM Group Corporation Limited (GRINM), No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China.,GRIMAT Engineering Institute Co., Ltd, No. 11, Xingke East St., Yanqi Economic Development Zone, Huairou District, Beijing 101407, P.R. China.,General Research Institute for Nonferrous Metals, No. 2, XinJieKouWai St., HaiDian District, Beijing 100088, P.R. China
| | - Bin Ma
- School of Basic Medical Sciences, Evidence-Based Medicine Center, Lanzhou University, No 199, Donggang West Road, Chengguan District, Lanzhou 730000, P. R. China
| |
Collapse
|
27
|
Chen Z, Zhao F, Liang C, Hu L, Li D, Zhang Y, Yin C, Chen L, Wang L, Lin X, Su P, Ma J, Yang C, Tian Y, Zhang W, Li Y, Peng S, Chen W, Zhang G, Qian A. Silencing of miR-138-5p sensitizes bone anabolic action to mechanical stimuli. Theranostics 2020; 10:12263-12278. [PMID: 33204341 PMCID: PMC7667683 DOI: 10.7150/thno.53009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Emerging evidence is revealing that microRNAs (miRNAs) play essential roles in mechanosensing for regulating osteogenesis. However, no mechanoresponsive miRNAs have been identified in human bone specimens. Methods: Bedridden and aged patients, hindlimb unloaded and aged mice, and Random Positioning Machine and primary aged osteoblasts were adopted to simulate mechanical unloading conditions at the human, animal and cellular levels, respectively. Treadmill exercise and Flexcell cyclic mechanical stretching were used to simulate mechanical loading in vivo and in vitro, respectively. Results: Here, we found increased miR-138-5p levels with a lower degree of bone formation in bone specimens from bedridden and aged patients. Loss- and gain-of-function studies showed that miR-138-5p directly targeted microtubule actin crosslinking factor 1 (MACF1) to inhibit osteoblast differentiation under different mechanical conditions. Regarding translational medicine, bone-targeted inhibition of miR-138-5p attenuated the decrease in the mechanical bone anabolic response in hindlimb unloaded mice. Moreover, bone-targeted inhibition of miR-138-5p sensitized the bone anabolic response to mechanical loading in both miR-138-5p transgenic mice and aged mice to promote bone formation. Conclusion: These data suggest that miR-138-5p as a mechanoresponsive miRNA accounts for the mechanosensitivity of the bone anabolic response and that inhibition of miR-138-5p in osteoblasts may be a novel bone anabolic sensitization strategy for ameliorating disuse or senile osteoporosis.
Collapse
|
28
|
He T, Liu W, Cao L, Liu Y, Zou Z, Zhong Y, Wang H, Mo Y, Peng S, Shuai C. CircRNAs and LncRNAs in Osteoporosis. Differentiation 2020; 116:16-25. [PMID: 33157509 DOI: 10.1016/j.diff.2020.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/16/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023]
Abstract
Osteoporosis is a systemic bone disease with bone fragility and increased fracture risk. The non-coding RNAs (ncRNAs) have appeared as important regulators of cellular signaling and pertinent human diseases. Studies have demonstrated that circular RNAs (circRNAs) and long non-coding RNAs (lncRNAs) are involved in the progression of osteoporosis through a variety of pathways, and are considered as targets for the prophylaxis and treatment of osteoporosis. Based on an in-depth understanding of their roles and mechanisms in osteoporosis, we summarize the functions and molecular mechanisms of circRNAs and lncRNAs involved in the progression of osteoporosis and provide some new insights for the prognosis, diagnosis and treatment of osteoporosis.
Collapse
Affiliation(s)
- Tiantian He
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Institute of Metabolism and Endocrinology, The Second Xiang-Ya Hospital, Central South University, 410011, Changsha, Hunan, People's Republic of China
| | - Lihua Cao
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Liu
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zi Zou
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yancheng Zhong
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haihua Wang
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuqing Mo
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuping Peng
- NHC Key Laboratory of Carcinogenesis of Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Non Resolving Inflammation and Cancer, Disease Genome Research Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Cijun Shuai
- Jiangxi University of Science and Technology, Ganzhou, 341000, China; State Key Laboratory of High Performance Complex Manufacturing, Central South University, Changsha, 410083, China.
| |
Collapse
|
29
|
Zhang L, Liu Y, Feng B, Liu LG, Zhou YC, Tang H. MiR-138-5p knockdown promotes osteogenic differentiation through FOXC1 up-regulation in human bone mesenchymal stem cells. Biochem Cell Biol 2020; 99:296-303. [PMID: 33058690 DOI: 10.1139/bcb-2020-0163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study examined the hypothesis that the microRNA miR-138-5p reduces the osteodifferentiation of human bone mesenchymal stem cells (hBMSCs) by downregulating the expression of forkhead box C1 (FOXC1). For this, hBMSCs were separated from bone marrow and osteogenic induction medium was added to stimulate osteogenic differentiation. Flow cytometric analysis was applied to evaluate the expression of cell-surface antigens associated with hBMSCs, including CD29, CD44, CD90, CD45, and CD34. qRT-PCR assays and Western blot assays were used to measure the mRNA and protein expression of miR-138-5p, osteocalcin, runt-related transcription factor 2, bone sialoprotein, alkaline phosphatase (ALP), and FOXC1. ALP staining assays and Alizarin Red staining (ARS) assays were used to confirm osteogenic differentiation. We used a luciferase assay to test the interaction between miR-138-5p and FOXC1. We demonstrated that miR-138-5p is downregulated in osteogenic differentiated hBMSCs. Further, overexpression of miR-138-5p reduced the expression of markers for osteodifferentiation, ALP activity, and ARS activity. Furthermore, we showed that FOXC1 is a downstream target gene of miR-138-5p, and that knockdown of miR-138-5p improves the osteogenesis differentiation of hBMSCs by upregulating FOXC1. The results from this study indicate miR-138-5p as a new target for osteogenic differentiation of hBMSCs and the treatment of bone defects.
Collapse
Affiliation(s)
- Lan Zhang
- Department of Orthopedics, Beijing Friendship Hospital Capital Medical University, Beijing 100050, P.R. China
| | - Yan Liu
- Department of Orthopedics, The Third Clinical Medical College of Inner Mongolia Medical University, Baotou 014010, P.R. China
| | - Bo Feng
- Department of Hand, Foot and Ankle Surgery, The Third Clinical Medical College of Inner Mongolia Medical University, Baotou 014010, P.R. China
| | - Li-Gong Liu
- Department of Hand, Foot and Ankle Surgery, The Third Clinical Medical College of Inner Mongolia Medical University, Baotou 014010, P.R. China
| | - Ying-Cai Zhou
- Department of Hand, Foot and Ankle Surgery, The Third Clinical Medical College of Inner Mongolia Medical University, Baotou 014010, P.R. China
| | - Hai Tang
- Department of Orthopedics, Beijing Friendship Hospital Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
30
|
Wang Z, Liu Q, Liu C, Tan W, Tang M, Zhou X, Sun T, Deng Y. Mg 2+ in β-TCP/Mg-Zn composite enhances the differentiation of human bone marrow stromal cells into osteoblasts through MAPK-regulated Runx2/Osx. J Cell Physiol 2020; 235:5182-5191. [PMID: 31742679 DOI: 10.1002/jcp.29395] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/27/2019] [Indexed: 12/15/2022]
Abstract
Inducing the osteogenic differentiation from bone marrow stromal cells (BMSCs) might be a potent strategy for treating bone loss and nonunion during fracture and improving fracture healing. Among several signaling pathways involved, mitogen-activated protein kinases (MAPKs) have been reported to play a critical role. Magnesium (Mg)-based alloys, including Mg-Zn alloy, have been used clinically as implants in the musculoskeletal field and could promote BMSC osteogenic differentiation. However, the underlying mechanisms remain unclear. In this study, we produced Mg-Zn alloy consists of Mg and low concentrations of Zn, calcium carbonate, and β-tricalcium phosphate (β-TCP; manifesting process not shown), prepared Mg, Zn, and Mg-Zn extracts, and investigated the specific effects of these extracts on human BMSC (hBMSC) osteogenic differentiation and MAPK signaling. Mg extracts and Mg-Zn extracts could significantly promote the osteogenic differentiation of hBMSCs as manifested as increased alkaline phosphatase levels, enhanced calcium nodules formation, and increased messenger RNA expression and protein levels of osteogenesis markers, including BMPs, Col-I, Runx2, and Osx; in the meantime, Mg culture medium (CM) and Mg-Zn CM both significantly enhanced the activation of MAPK signaling in hBMSCs. By adding ERK1/2 signaling, p38 signaling, or JNK signaling inhibitor to Mg-Zn CM, or conducting p38 MAPK silence in hBMSCs, we revealed that these extracts might promote hBMSC osteogenic differentiation via p38 MAPK signaling and MAPK-regulated Runx2/Osx. In conclusion, Mg2+ in β-TCP/Mg-Zn extract promotes the osteogenic differentiation of hBMSCs via MAPK-regulated Runx2/Osx interaction.
Collapse
Affiliation(s)
- Zhenting Wang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Urology Surgery, Haikou People's Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Qing Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Congcong Liu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Wei Tan
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| | - Mingying Tang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaohua Zhou
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tianshi Sun
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youwen Deng
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan
| |
Collapse
|