1
|
Singh M, Singh P, Singh B, Sharma K, Kumar N, Singh D, Mastana S. Molecular Signaling Pathways and MicroRNAs in Bone Remodeling: A Narrative Review. Diseases 2024; 12:252. [PMID: 39452495 PMCID: PMC11507001 DOI: 10.3390/diseases12100252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
Bone remodeling is an intricate process executed throughout one's whole life via the cross-talk of several cellular events, progenitor cells and signaling pathways. It is an imperative mechanism for regaining bone loss, recovering damaged tissue and repairing fractures. To achieve this, molecular signaling pathways play a central role in regulating pathological and causal mechanisms in different diseases. Similarly, microRNAs (miRNAs) have shown promising results in disease management by mediating mRNA targeted gene expression and post-transcriptional gene function. However, the role and relevance of these miRNAs in signaling processes, which regulate the delicate balance between bone formation and bone resorption, are unclear. This review aims to summarize current knowledge of bone remodeling from two perspectives: firstly, we outline the modus operandi of five major molecular signaling pathways, i.e.,the receptor activator of nuclear factor kappa-B (RANK)-osteoprotegrin (OPG) and RANK ligand (RANK-OPG-RANKL), macrophage colony-stimulating factor (M-CSF), Wnt/β-catenin, Jagged/Notch and bone morphogenetic protein (BMP) pathways in regards to bone cell formation and function; and secondly, the miRNAs that participate in these pathways are introduced. Probing the miRNA-mediated regulation of these pathways may help in preparing the foundation for developing targeted strategies in bone remodeling, repair and regeneration.
Collapse
Affiliation(s)
- Monica Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Puneetpal Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Baani Singh
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Kirti Sharma
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Nitin Kumar
- Department of Human Genetics, Punjabi University, Patiala 147002, India; (M.S.); (B.S.); (K.S.); (N.K.)
| | - Deepinder Singh
- VardhmanMahavir Health Care, Urban Estate Ph-II, Patiala 147002, India;
| | - Sarabjit Mastana
- Human Genomics Laboratory, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough LE11 3TU, UK;
| |
Collapse
|
2
|
Castaño IM, Raftery RM, Chen G, Cavanagh B, Quinn B, Duffy GP, Curtin CM, O'Brien FJ. Dual scaffold delivery of miR-210 mimic and miR-16 inhibitor enhances angiogenesis and osteogenesis to accelerate bone healing. Acta Biomater 2023; 172:480-493. [PMID: 37797708 DOI: 10.1016/j.actbio.2023.09.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/30/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023]
Abstract
Angiogenesis is critical for successful bone repair, and interestingly, miR-210 and miR-16 possess counter-active targets involved in both angiogenesis and osteogenesis: miR-210 acts as an activator by silencing EFNA3 & AcvR1b, while miR-16 inhibits both pathways by silencing VEGF & Smad5. It was thus hypothesized that dual delivery of both a miR-210 mimic and a miR-16 inhibitor from a collagen-nanohydroxyapatite scaffold system may hold significant potential for bone repair. Therefore, this systems potential to rapidly accelerate bone repair by directing enhanced angiogenic-osteogenic coupling in host cells in a rat calvarial defect model at a very early 4 week timepoint was assessed. In vitro, the treatment significantly enhanced angiogenic-osteogenic coupling of human mesenchymal stem cells, with enhanced calcium deposition after just 10 days in 2D and 14 days on scaffolds. In vivo, these dual-miRNA loaded scaffolds showed more than double bone volume and vessel recruitment increased 2.3 fold over the miRNA-free scaffolds. Overall, this study demonstrates the successful development of a dual-miRNA mimic/inhibitor scaffold for enhanced in vivo bone repair for the first time, and the possibility of extending this 'off-the-shelf' platform system to applications beyond bone offers immense potential to impact a myriad of other tissue engineering areas. STATEMENT OF SIGNIFICANCE: miRNAs have potential as a new class of bone healing therapeutics as they can enhance the regenerative capacity of bone-forming cells. However, angiogenic-osteogenic coupling is critical for successful bone repair. Therefore, this study harnesses the delivery of miR-210, known to be an activator of both angiogenesis and osteogenesis, and miR-16 inhibitor, as miR-16 is known to inhibit both pathways, from a collagen-nanohydroxyapatite scaffold system to rapidly enhance osteogenesis in vitro and bone repair in vivo in a rat calvarial defect model. Overall, it describes the successful development of the first dual-miRNA mimic/inhibitor scaffold for enhanced in vivo bone repair. This 'off-the-shelf' platform system offers immense potential to extend beyond bone applications and impact a myriad of other tissue engineering areas.
Collapse
Affiliation(s)
- Irene Mencía Castaño
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland
| | - Rosanne M Raftery
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland; School of Pharmacy, RCSI, Dublin, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility, RCSI, Dublin 2, Ireland
| | | | - Brian Quinn
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland
| | - Garry P Duffy
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin 2, Ireland; Anatomy, School of Medicine, College of Medicine Nursing and Health Sciences, University of Galway, University Road, Galway, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin 2, Ireland.
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin 2, Ireland.
| |
Collapse
|
3
|
Shams R, Behmanesh A, Mazhar FN, Vaghari AA, Hossein-Khannazer N, Agarwal T, Vosough M, Padrón JM. Developed Bone Biomaterials Incorporated with MicroRNAs to Promote Bone Regeneration: A Systematic Review, Bioinformatics, and Meta-analysis Study. ACS Biomater Sci Eng 2023; 9:5186-5204. [PMID: 37585807 DOI: 10.1021/acsbiomaterials.3c00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
This systematic review and meta-analysis focused on the effectiveness of biomaterials integrated with specific microRNAs (miRNAs) for bone fracture repair treatment. We conducted a comprehensive search of the PubMed, Web of Science, and Scopus databases, identifying 42 relevant papers up to March 2022. Hydrogel-based scaffolds were the most commonly used, incorporating miRNAs like miR-26a, miR-21, and miR-222, with miR-26a being the most prevalent. The meta-analysis revealed significant benefits of incorporating miRNAs into scaffolds for bone repair, particularly in hydrogel scaffolds. However, some controversies were observed among studies, presenting challenges in selecting appropriate miRNAs for this purpose. The study concludes that incorporating specific miRNAs into bone biomaterials enhances bone regeneration, but further trials comparing different biomaterials and miRNAs are necessary to validate their potential applications for bone tissue regeneration.
Collapse
Affiliation(s)
- Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Ali Behmanesh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Farid Najd Mazhar
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Amir Ali Vaghari
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Tarun Agarwal
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh 522302, India
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 171 77 Stockholm, Sweden
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, P.O. Box 456, 38200 La Laguna, Spain
| |
Collapse
|
4
|
Su D, Swearson S, Krongbaramee T, Sun H, Hong L, Amendt BA. Exploring microRNAs in craniofacial regenerative medicine. Biochem Soc Trans 2023; 51:841-854. [PMID: 37073783 PMCID: PMC11244734 DOI: 10.1042/bst20221448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023]
Abstract
microRNAs (miRs) have been reported over the decades as important regulators in bone development and bone regeneration. They play important roles in maintaining the stem cell signature as well as regulating stem cell fate decisions. Thus, delivering miRs and miR inhibitors to the defect site is a potential treatment towards craniofacial bone defects. However, there are challenges in translation of basic research to clinics, including the efficiency, specificity, and efficacy of miR manipulation methods and the safety of miR delivery systems. In this review, we will compare miR oligonucleotides, mimics and antagomirs as therapeutic reagents to treat disease and regenerate tissues. Newer technology will be discussed as well as the efficiency and efficacy of using these technologies to express or inhibit miRs in treating and repairing oral tissues. Delivery of these molecules using extracellular vesicles and nanoparticles can achieve different results and depending on their composition will elicit specific effects. We will highlight the specificity, toxicity, stability, and effectiveness of several miR systems in regenerative medicine.
Collapse
Affiliation(s)
- Dan Su
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
| | - Samuel Swearson
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
| | - Tadkamol Krongbaramee
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
- Division of Endodontics, Department of Restorative Dentistry & Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Hongli Sun
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Liu Hong
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| | - Brad A Amendt
- Department of Anatomy and Cell Biology, The University of Iowa, Iowa City, IA, U.S.A
- Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, U.S.A
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, U.S.A
| |
Collapse
|
5
|
Camacho-Alonso F, Tudela-Mulero MR, Buendía AJ, Navarro JA, Pérez-Sayáns M, Mercado-Díaz AM. Bone regeneration in critical-sized mandibular symphysis defects using bioceramics with or without bone marrow mesenchymal stem cells in healthy, diabetic, osteoporotic, and diabetic-osteoporotic rats. Dent Mater 2022; 38:1283-1300. [PMID: 35717229 DOI: 10.1016/j.dental.2022.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/14/2022] [Accepted: 06/05/2022] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To compare new bone formation in mandibular critical-sized bone defects (CSBDs) in healthy, diabetic, osteoporotic, and diabetic-osteoporotic rats filled with bioceramics (BCs) with or without bone marrow mesenchymal stem cells (BMSCs). METHODS A total of 64 adult female Sprague-Dawley rats were randomized into four groups (n = 16 per group): Group 1 healthy, Group 2 diabetic, Group 3 osteoporotic, and Group 4 diabetic-osteoporotic rats. Streptozotocin was used to induce type 1 diabetes in Group 2 and 4, while bilateral ovariectomy was used to induce osteoporosis in Group 3 and 4. The central portion of the rat mandibular symphysis was used as a physiological CSBD. In each group, eight defects were filled with BC (hydroxypatatite 60% and β-tricalcium phosphate 40%) alone and eight with BMSCs cultured on BC. The animals were sacrificed at 4 and 8 weeks, and the mandibles were processed for micro-computed tomography to analyze radiological union and bone mineral density (BMD); histological analysis of the bone union; and immunohistochemical analysis, which included immunoreactivity of vascular endothelial growth factor (VEGF) and bone morphogenetic protein 2 (BMP-2). RESULTS In all groups (healthy, diabetics, osteoporotics, and diabetics-osteoporotics), the CSBDs filled with BC + BMSCs showed greater radiological bone union, BMD, histological bone union, and more VEGF and BMP-2 positivity, in comparison with CSBDs treated with BC alone (at 4 and 8 weeks). CONCLUSIONS Application of BMSCs cultured on BCs improves bone regeneration in CSBDs compared with application of BCs alone in healthy, diabetic, osteoporotic, and diabetic-osteoporotic rats.
Collapse
Affiliation(s)
- F Camacho-Alonso
- Department of Oral Surgery, University of Murcia, Murcia, Spain.
| | | | - A J Buendía
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - J A Navarro
- Department of Histology and Pathological Anatomy, University of Murcia, Murcia, Spain
| | - M Pérez-Sayáns
- Department of Oral Medicine, Oral Surgery and Implantology, University of Santiago de Compostela, Spain. MedOralRes Group, Health Research Institute of Santiago de Compostela (IDIS). Santiago de Compostela, Spain
| | | |
Collapse
|
6
|
Epigenetic therapy targeting bone marrow mesenchymal stem cells for age-related bone diseases. Stem Cell Res Ther 2022; 13:201. [PMID: 35578312 PMCID: PMC9109405 DOI: 10.1186/s13287-022-02852-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/14/2022] [Indexed: 02/08/2023] Open
Abstract
As global aging accelerates, the prevention and treatment of age-related bone diseases are becoming a critical issue. In the process of senescence, bone marrow mesenchymal stem cells (BMSCs) gradually lose the capability of self-renewal and functional differentiation, resulting in impairment of bone tissue regeneration and disorder of bone tissue homeostasis. Alteration in epigenetic modification is an essential factor of BMSC dysfunction during aging. Its transferability and reversibility provide the possibility to combat BMSC aging by reversing age-related modifications. Emerging evidence demonstrates that epigenetic therapy based on aberrant epigenetic modifications could alleviate the senescence and dysfunction of stem cells. This review summarizes potential therapeutic targets for BMSC aging, introduces some potential approaches to alleviating BMSC aging, and analyzes its prospect in the clinical application of age-related bone diseases.
Collapse
|
7
|
Tevlek A, Topuz B, Akbay E, Aydin HM. Surface channel patterned and endothelialized poly(glycerol sebacate) based elastomers. J Biomater Appl 2022; 37:287-302. [DOI: 10.1177/08853282221085798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prevascularization of tissue equivalents is critical for fulfilling the need for sufficient vascular organization for nutrient and gas transport. Hence, endothelial cell culture on biomaterials is of great importance for researchers. Numerous alternate strategies have been suggested in this sense, with cell-based methods being the most commonly employed. In this study, poly (glycerol sebacate) (PGS) elastomers with varying crosslinking ratios were synthesized and their surfaces were patterned with channels by using laser ablation technique. In order to determine an ideal material for cell culture studies, the elastomers were subsequently mechanically, chemically, and biologically characterized. Following that, human umbilical vein endothelial cells (HUVECs) were seeded into the channels established on the PGS membranes and cultured under various culture conditions to establish the optimal culture parameters. Lastly, the endothelial cell responses to the synthesized PGS elastomers were evaluated. Remarkable cell proliferation and impressive cellular organizations were noticed on the constructs created as part of the investigation. On the concrete output of this research, arrangements in various geometries can be created by laser ablation method and the effects of various molecules, drugs or agents on endothelial cells can be evaluated. The platforms produced can be employed as an intermediate biomaterial layer containing endothelial cells for vascularization of tissue-engineered structures, particularly in layer-by-layer tissue engineering approaches.
Collapse
Affiliation(s)
- Atakan Tevlek
- Institute of Science, Bioengineering Division, Hacettepe University, Ankara, Turkey
| | - Bengisu Topuz
- Institute of Science, Bioengineering Division, Hacettepe University, Ankara, Turkey
| | - Esin Akbay
- Faculty of Science, Department of Biology, Hacettepe University, Ankara, Turkey
| | - Halil Murat Aydin
- Institute of Science, Bioengineering Division, Hacettepe University, Ankara, Turkey
- Centre for Bioengineering, Hacettepe University, Ankara, Turkey§Current Affiliation: METU MEMS Center, Ankara, Turkey
| |
Collapse
|
8
|
Groven RVM, van Koll J, Poeze M, Blokhuis TJ, van Griensven M. miRNAs Related to Different Processes of Fracture Healing: An Integrative Overview. Front Surg 2021; 8:786564. [PMID: 34869574 PMCID: PMC8639603 DOI: 10.3389/fsurg.2021.786564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 12/21/2022] Open
Abstract
Fracture healing is a complex, dynamic process that is directed by cellular communication and requires multiple cell types, such as osteoblasts, osteoclasts, and immune cells. Physiological fracture healing can be divided into several phases that consist of different processes, such as angiogenesis, osteogenesis, and bone resorption/remodelling. This is needed to guarantee proper bone regeneration after fracture. Communication and molecular regulation between different cell types and within cells is therefore key in successfully orchestrating these processes to ensure adequate bone healing. Among others, microRNAs (miRNAs) play an important role in cellular communication. microRNAs are small, non-coding RNA molecules of ~22 nucleotides long that can greatly influence gene expression by post-transcriptional regulation. Over the course of the past decade, more insights have been gained in the field of miRNAs and their role in cellular signalling in both inter- and intracellular pathways. The interplay between miRNAs and their mRNA targets, and the effect thereof on different processes and aspects within fracture healing, have shown to be interesting research topics with possible future diagnostic and therapeutic potential. Considering bone regeneration, research moreover focusses on specific microRNAs and their involvement in individual pathways. However, it is required to combine these data to gain more understanding on the effects of miRNAs in the dynamic process of fracture healing, and to enhance their translational application in research, as well as in the clinic. Therefore, this review aims to provide an integrative overview on miRNAs in fracture healing, related to several key aspects in the fracture healing cascade. A special focus will be put on hypoxia, angiogenesis, bone resorption, osteoclastogenesis, mineralization, osteogenesis, osteoblastogenesis, osteocytogenesis, and chondrogenesis.
Collapse
Affiliation(s)
- Rald V M Groven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands.,Division of Traumasurgery, Department of Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Johan van Koll
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Martijn Poeze
- Division of Traumasurgery, Department of Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Taco J Blokhuis
- Division of Traumasurgery, Department of Surgery, Maastricht University Medical Center, Maastricht, Netherlands
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
9
|
Kharaghani D, Kurniwan EB, Khan MQ, Yoshiko Y. MiRNA-Nanofiber, the Next Generation of Bioactive Scaffolds for Bone Regeneration: A Review. MICROMACHINES 2021; 12:mi12121472. [PMID: 34945325 PMCID: PMC8707075 DOI: 10.3390/mi12121472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 12/19/2022]
Abstract
Scaffold-based bone tissue engineering has been introduced as an alternative treatment option for bone grafting due to limitations in the allograft. Not only physical conditions but also biological conditions such as gene expression significantly impact bone regeneration. Scaffolds in composition with bioactive molecules such as miRNA mimics provide a platform to enhance migration, proliferation, and differentiation of osteoprogenitor cells for bone regeneration. Among scaffolds, fibrous structures showed significant advantages in promoting osteogenic differentiation and bone regeneration via delivering bioactive molecules over the past decade. Here, we reviewed the bone and bone fracture healing considerations for the impact of miRNAs on bone regeneration. We also examined the methods used to improve miRNA mimics uptake by cells, the fabrication of fibrous scaffolds, and the effective delivery of miRNA mimics using fibrous scaffold and their processes for bone development. Finally, we offer our view on the principal challenges of miRNA mimics delivery by nanofibers for bone tissue engineering.
Collapse
Affiliation(s)
- Davood Kharaghani
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
- Correspondence: ; Tel.: +81-82-257-5621
| | - Eben Bashir Kurniwan
- School of Dentistry, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
| | - Muhammad Qamar Khan
- Nanotechnology Research Lab, Department of Textile and Clothing, National Textile University, Karachi Campus, Karachi 74900, Pakistan;
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan;
| |
Collapse
|
10
|
Alfotawi R, Ahmed R, Atteya M, Mahmood A, Siyal A, AlHindi M, El-Ghannam A. Assessment of novel surgical procedures using decellularised muscle and bioactive ceramic: a histological analysis. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:113. [PMID: 34453610 PMCID: PMC8403111 DOI: 10.1007/s10856-021-06585-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 06/21/2021] [Indexed: 06/13/2023]
Abstract
Tissue regeneration and neovascularisation in cases of major bone loss is a challenge in maxillofacial surgery. The hypothesis of the present study is that the addition of resorbable bioactive ceramic Silica Calcium Phosphate Cement (SCPC) to Declluraized Muscle Scaffold (DSM) can expedite bone formation and maturation. Two surgical defect models were created in 18 nude transgenic mice. Group 1(n = 6), with a 2-mm decortication calvarial defect, was treated with a DSM/SCPC sheet over the corticated bone as an onlay then seeded with human Mesenchymal Stromal Cells hMSC in situ. In Group 2 (n = 6), a critical size (4 mm) calvarial defect was made and grafted with DSM/SCPC/in situ human bone marrow stromal cells (hMSCs). The control groups included Group 3 (n = 3) animals, with a 2-mm decortication defect treated with an onlay DSM sheet, and Group 4 (n = 3) animals, treated with critical size defect grafted with plain DSM. After 8 weeks, bone regeneration in various groups was evaluated using histology, immunohistochemistry and histomorphometry. New bone formation and maturation was superior in groups treated with DSM/SCPC/hMSC. The DMS/SCPC scaffold has the ability to augment and induce bone regeneration and neovascularisation in cases of major bone resorption and critical size defects.
Collapse
Affiliation(s)
- Randa Alfotawi
- Oral & Maxillofacial dept, Dental Collage, King Saud University, Riyadh, Saudi Arabia.
| | - Raeesa Ahmed
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Atteya
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Amer Mahmood
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Marium AlHindi
- Oral & Maxillofacial dept, Dental Collage, King Saud University, Riyadh, Saudi Arabia
| | - Ahmad El-Ghannam
- Department of Mechanical Engineering and Engineering Science, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
11
|
Wang G, Wan L, Zhang L, Yan C, Zhang Y. MicroRNA-133a Regulates the Viability and Differentiation Fate of Bone Marrow Mesenchymal Stem Cells via MAPK/ERK Signaling Pathway by Targeting FGFR1. DNA Cell Biol 2021; 40:1112-1123. [PMID: 34165368 DOI: 10.1089/dna.2021.0206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dysfunction of bone marrow mesenchymal stem cells (BMSCs) is recognized critical in bone deteriorations of osteoporosis. However, the specific mechanisms that determine the fate of BMSCs remain elusive. MicroRNA-133a (miR-133a), a highly conserved microRNA, was investigated under both in vitro and in vivo conditions. In the in vitro study, cell proliferation, cell apoptosis, and osteoblast/adipocyte differentiation of BMSCs as a result of overexpression or knockdown of miR-133a was investigated. In the in vivo study, the ovariectomy (OVX) model was applied on mice, with further treatment of the models with BMSC-specific miR-133a antagomir through femur intramedullary injection. Microcomputed tomography scanning and histological analysis of the proximal and middle femur were performed to evaluate the morphological changes. The results revealed that overexpression of miR-133a suppressed cell proliferation, cell viability, and osteoblast differentiation of BMSCs, but increased adipocyte differentiation. We also found that FGFR1, an important upstream regulator of mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signal pathway, was a major target of miR-133a. We also recorded that BMSC-specific knockdown of miR-133a attenuates bone loss in OVX mice. Our study suggested that miR-133a played an important role in maintaining the viability and balance between osteoblast and adipocyte differentiation of BMSCs through the MAPK/ERK signaling pathway by targeting FGFR1.
Collapse
Affiliation(s)
- Gang Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lifu Wan
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lecheng Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chao Yan
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuelei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
12
|
Wu Z, Jin K, Wang L, Fan Y. A Review: Optimization for Poly(glycerol sebacate) and Fabrication Techniques for Its Centered Scaffolds. Macromol Biosci 2021; 21:e2100022. [PMID: 34117837 DOI: 10.1002/mabi.202100022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/26/2021] [Indexed: 12/29/2022]
Abstract
Poly(glycerol sebacate) (PGS), an emerging promising thermosetting polymer synthesized from sebacic acid and glycerol, has attracted considerable attention due to its elasticity, biocompatibility, and tunable biodegradation properties. But it also has some drawbacks such as harsh synthesis conditions, rapid degradation rates, and low stiffness. To overcome these challenges and optimize PGS performance, various modification methods and fabrication techniques for PGS-based scaffolds have been developed in recent years. Outlining the current modification approaches of PGS and summarizing the fabrication techniques for PGS-based scaffolds are of great importance to accelerate the development of new materials and enable them to be appropriately used in potential applications. Thus, this review comprehensively overviews PGS derivatives, PGS composites, PGS blends, processing for PGS-based scaffolds, and their related applications. It is envisioned that this review could instruct and inspire the design of the PGS-based materials and facilitate tissue engineering advances into clinical practice.
Collapse
Affiliation(s)
- Zebin Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Kaixiang Jin
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Lizhen Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.,School of Medical Science and Engineering, Beihang University, Beijing, 100083, China
| |
Collapse
|
13
|
Li L, Peng Y, Yuan Q, Sun J, Zhuang A, Bi X. Cathelicidin LL37 Promotes Osteogenic Differentiation in vitro and Bone Regeneration in vivo. Front Bioeng Biotechnol 2021; 9:638494. [PMID: 34012955 PMCID: PMC8126666 DOI: 10.3389/fbioe.2021.638494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 04/08/2021] [Indexed: 11/17/2022] Open
Abstract
Different types of biomaterials have been used to repair the defect of bony orbit. However, exposure and infections are still critical risks in clinical application. Biomaterials with characteristics of osteogenesis and antibiosis are needed for bone regeneration. In this study, we aimed to characterize the antimicrobial effects of cathelicidin-LL37 and to assess any impacts on osteogenic activity. Furthermore, we attempted to demonstrate the feasibility of LL37 as a potential strategy in the reconstruction of clinical bone defects. Human adipose-derived mesenchyme stem cells (hADSCs) were cultured with different concentrations of LL37 and the optimum concentration for osteogenesis was selected for further in vitro studies. We then evaluated the antibiotic properties of LL37 at the optimum osteogenic concentration. Finally, we estimated the efficiency of a PSeD/hADSCs/LL37 combined scaffold on reconstructing bone defects in the rat calvarial defect model. The osteogenic ability on hADSCs in vitro was shown to be dependent on the concentration of LL37 and reached a peak at 4 μg/ml. The optimum concentration of LL37 showed good antimicrobial properties against Escherichia coli and Staphylococcus anurans. The combination scaffold of PSeD/hADSCs/LL37 showed superior osteogenic properties compared to the PSeD/hADSCs, PSeD, and control groups scaffolds, indicating a strong bone reconstruction effect in the rat calvarial bone defect model. In Conclusion, LL37 was shown to promote osteogenic differentiation in vitro as well as antibacterial properties. The combination of PSeD/hADSCs/LL37 was advantageous in the rat calvarial defect reconstruction model, showing high potential in clinical bone regeneration.
Collapse
Affiliation(s)
- Lunhao Li
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yiyu Peng
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Qingyue Yuan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Jing Sun
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Ai Zhuang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xiaoping Bi
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
14
|
Hong L, Sun H, Amendt BA. MicroRNA function in craniofacial bone formation, regeneration and repair. Bone 2021; 144:115789. [PMID: 33309989 PMCID: PMC7869528 DOI: 10.1016/j.bone.2020.115789] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
Bone formation in the craniofacial complex is regulated by cranial neural crest (CNC) and mesoderm-derived cells. Different elements of the developing skull, face, mandible, maxilla (jaws) and nasal bones are regulated by an array of transcription factors, signaling molecules and microRNAs (miRs). miRs are molecular modulators of these factors and act to restrict their expression in a temporal-spatial mechanism. miRs control the different genetic pathways that form the craniofacial complex. By understanding how miRs function in vivo during development they can be adapted to regenerate and repair craniofacial genetic anomalies as well as bone diseases and defects due to traumatic injuries. This review will highlight some of the new miR technologies and functions that form new bone or inhibit bone regeneration.
Collapse
Affiliation(s)
- Liu Hong
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, USA
| | - Hongli Sun
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, USA
| | - Brad A Amendt
- Iowa Institute for Oral Health Research, The University of Iowa, Iowa City, IA, USA; The University of Iowa, Department of Anatomy and Cell Biology, Iowa City, IA, USA; Craniofacial Anomalies Research Center, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
15
|
Chen M, Zhou M, Fu Y, Li J, Wang Z. Effects of miR-672 on the angiogenesis of adipose-derived mesenchymal stem cells during bone regeneration. Stem Cell Res Ther 2021; 12:85. [PMID: 33494825 PMCID: PMC7836178 DOI: 10.1186/s13287-021-02154-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/07/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Sufficient vascular network plays an important role in the repair of bone defects. Bone morphogenetic protein 2 (BMP2) being a key regulator of angiogenesis has attracted the attention of researchers. In addition, evidence has suggested that BMP2 coordinates with microRNAs (miRNAs) to form intracellular networks regulating mesenchymal stem cells (MSCs) angiogenesis. Elucidating the underlying mechanisms that are regulating adipose-derived mesenchymal stem cells (ADSCs) angiogenesis might provide more effective method to enhance bone regeneration. METHODS We identified the specific miRNA in rat ADSCs during BMP2-induced angiogenesis and chose the most significant differentially expressed miRNA, miR-672. Three lentiviral system named Lenti-miR-672, Lenti-as-miR-672, and Lenti-miR-NC were transduced into the ADSCs individually. Then, the quantitative real-time polymerase chain reaction (qPCR), western blotting, and blood vessel formation analysis were performed to investigate the effects of miR-672 on ADSCs angiogenesis. Bioinformation platforms were used to screen the potential target of miR-672. Small interfering RNA (siRNA) against TIMP2 (si-TIMP2) mRNA were obtained from GenePharma, and then si-TIMP2 miRNA and miR-672 were co-transfected into ADSCs to detect the effects of TIMP2 on angiogenesis. Calcium phosphate cement (CPC) scaffolds that seeded the lentiviral-modified ADSCs were constructed to test the vascularized bone regeneration in vivo. RESULTS Our data showed that after the angiogenesis of ADSCs induced by BMP2, miR-672 was the most significantly upregulated miRNA. Overexpression of miR-672 promoted the angiogenesis of ADSCs, while knockdown of miR-672 repressed the angiogenesis of ADSCs. The bioinformation prediction showed that TIMP2 might be the one of miR-672' potential targets. TIMP2 protein expression was gradually decreased in ADSCs with overexpressed miR-672. And the angiogenic factors were upregulated in the ADSCs which were transduced with si-TIMP2. Then, the CPC scaffolds coupled the miR-672-modified ADSCs and showed the good potential in vascularized bone regeneration. The overexpressed miR-672 could greatly enhance the blood vessel volume and Microfil-labeled blood vessel numbers in newly formed bone. CONCLUSION BMP2 could promote the angiogenesis of ADSCs through stimulating the expression of miR-672 in ADSCs. miR-672 acted as a positive regulator on the angiogenesis of ADSCs, and incorporating the miR-672-modified ADSCs in the CPC could significantly promote the vascularization and the bone regeneration.
Collapse
Affiliation(s)
- Mingjiao Chen
- grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No. 639, Shanghai, 200011 People’s Republic of China
| | - Meng Zhou
- grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No. 639, Shanghai, 200011 People’s Republic of China
| | - Yao Fu
- grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No. 639, Shanghai, 200011 People’s Republic of China
| | - Jin Li
- grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No. 639, Shanghai, 200011 People’s Republic of China
| | - Zi Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Zhizaoju Road No. 639, Shanghai, 200011 People’s Republic of China
| |
Collapse
|
16
|
Shang F, Yu Y, Liu S, Ming L, Zhang Y, Zhou Z, Zhao J, Jin Y. Advancing application of mesenchymal stem cell-based bone tissue regeneration. Bioact Mater 2020; 6:666-683. [PMID: 33005830 PMCID: PMC7509590 DOI: 10.1016/j.bioactmat.2020.08.014] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/07/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022] Open
Abstract
Reconstruction of bone defects, especially the critical-sized defects, with mechanical integrity to the skeleton is important for a patient's rehabilitation, however, it still remains challenge. Utilizing biomaterials of human origin bone tissue for therapeutic purposes has provided a facilitated approach that closely mimics the critical aspects of natural bone tissue with regard to its properties. However, not only efficacious and safe but also cost-effective and convenient are important for regenerative biomaterials to achieve clinical translation and commercial success. Advances in our understanding of regenerative biomaterials and their roles in new bone formation potentially opened a new frontier in the fast-growing field of regenerative medicine. Taking inspiration from the role and multicomponent construction of native extracellular matrix (ECM) for cell accommodation, the ECM-mimicking biomaterials and the naturally decellularized ECM scaffolds were used to create new tissues for bone restoration. On the other hand, with the going deep in understanding of mesenchymal stem cells (MSCs), they have shown great promise to jumpstart and facilitate bone healing even in diseased microenvironments with pharmacology-based endogenous MSCs rescue/mobilization, systemic/local infusion of MSCs for cytotherapy, biomaterials-based approaches, cell-sheets/-aggregates technology and usage of subcellular vesicles of MSCs to achieve scaffolds-free or cell-free delivery system, all of them have been shown can improve MSCs-mediated regeneration in preclinical studies and several clinical trials. Here, following an overview discussed autogenous/allogenic and ECM-based bone biomaterials for reconstructive surgery and applications of MSCs-mediated bone healing and tissue engineering to further offer principles and effective strategies to optimize MSCs-based bone regeneration. Focusing on MSCs based bone regeneration. Discussed cytotherapy, cell-free therapies and cell-aggregates technology in detail. Stating the approaches of MSCs in diseased microenvironments.
Collapse
Affiliation(s)
- Fengqing Shang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Stomatology, The 306th Hospital of PLA, Beijing, 100101, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, Shandong, 250012, China
| | - Shiyu Liu
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Leiguo Ming
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yongjie Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhifei Zhou
- Department of Stomatology, General Hospital of Tibetan Military Command, Lhasa, 850000, China
| | - Jiayu Zhao
- Bureau of Service for Veteran Cadres of PLA in Beijing, Beijing, 100001, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research, Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Corresponding author.
| |
Collapse
|
17
|
Epigenetic Regulation in Mesenchymal Stem Cell Aging and Differentiation and Osteoporosis. Stem Cells Int 2020; 2020:8836258. [PMID: 32963550 PMCID: PMC7501554 DOI: 10.1155/2020/8836258] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a reliable source for cell-based regenerative medicine owing to their multipotency and biological functions. However, aging-induced systemic homeostasis disorders in vivo and cell culture passaging in vitro induce a functional decline of MSCs, switching MSCs to a senescent status with impaired self-renewal capacity and biased differentiation tendency. MSC functional decline accounts for the pathogenesis of many diseases and, more importantly, limits the large-scale applications of MSCs in regenerative medicine. Growing evidence implies that epigenetic mechanisms are a critical regulator of the differentiation programs for cell fate and are subject to changes during aging. Thus, we here review epigenetic dysregulations that contribute to MSC aging and osteoporosis. Comprehending detailed epigenetic mechanisms could provide us with a novel horizon for dissecting MSC-related pathogenesis and further optimizing MSC-mediated regenerative therapies.
Collapse
|
18
|
Castaño IM, Raftery RM, Chen G, Cavanagh B, Quinn B, Duffy GP, O'Brien FJ, Curtin CM. Rapid bone repair with the recruitment of CD206 +M2-like macrophages using non-viral scaffold-mediated miR-133a inhibition of host cells. Acta Biomater 2020; 109:267-279. [PMID: 32251781 DOI: 10.1016/j.actbio.2020.03.042] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/10/2020] [Accepted: 03/27/2020] [Indexed: 01/01/2023]
Abstract
microRNAs offer vast therapeutic potential for multiple disciplines. From a bone perspective, inhibition of miR-133a may offer potential to enhance Runx2 activity and increase bone repair. This study aims to assess the therapeutic capability of antagomiR-133a delivery from collagen-nanohydroxyapatite (coll-nHA) scaffolds following cell-free implantation in rat calvarial defects (7 mm diameter). This is, to the best of our knowledge, the first report of successful in vivo antagomiR uptake in host cells of fully immunocompetent animals without distribution to other off-target tissues. Our results demonstrate the localized release of antagomiR-133a to the implant site at 1 week post-implantation with increased calcium deposits already evident in the antagomiR-133a loaded scaffolds at this early timepoint. This was followed by an approximate 2-fold increase in bone volume versus antagomiR-free scaffolds and a significant 10-fold increase over the empty defect controls, after just 4 weeks. An increase in host CD206+ cells suggests an accelerated pro-remodeling response by M2-like macrophages accompanying bone repair with this treatment. Overall, this non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the addition of exogenous cells - and underlines the role of M2 macrophage-like cells in directing accelerated bone repair. Expanding the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a variety of therapeutic indications. STATEMENT OF SIGNIFICANCE: microRNAs, small non-coding RNA molecules involved in gene regulation, may have potential as a new class of bone healing therapeutics as they can enhance the regenerative capacity of bone-forming cells. We developed a collagen-nanohydroxyapatite-microRNA scaffold system to investigate whether miR133a inhibition can enhance osteogenesis in rat MSCs and ultimately accelerate endogenous bone repair by host cells in vivo without pre-seeding cells prior to implantation. Overall, this off-the-shelf, non-viral scaffold-mediated antagomiR-133a delivery platform demonstrates capability to accelerate bone repair in vivo - without the requirement of exogenous cells - and highlights the role of CD206+M2 macrophage-like cells in guiding accelerated bone repair. Translating the repertoire of this platform to deliver alternative miRNAs offers exciting possibilities for a vast myriad of therapeutic indications.
Collapse
|
19
|
Peng Y, Li L, Yuan Q, Gu P, You Z, Zhuang A, Bi X. Effect of Bifunctional β Defensin 2-Modified Scaffold on Bone Defect Reconstruction. ACS OMEGA 2020; 5:4302-4312. [PMID: 32149260 PMCID: PMC7057706 DOI: 10.1021/acsomega.9b04249] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/07/2020] [Indexed: 05/05/2023]
Abstract
Bone tissue engineering has emerged as an effective alternative treatment to the problem of bone defect. To repair a bone defect, antibiosis and osteogenesis are two essential aspects of the repair process. By searching the literature and performing exploratory experiments, we found that β defensin 2 (BD2), with bifunctional properties of antibiosis and osteogenesis, was a feasible alternative for traditional growth factors. The antimicrobial ability of BD2 against Staphylococcus aureus and Escherichia coli was studied by the spread plate and live/dead staining methods (low effective concentration of 20 ng/mL). BD2 was also demonstrated to enhance osteogenesis, with higher messenger RNA (mRNA) and protein expression of the osteogenic markers collagen I (Col1), runt-related transcription factor 2 (Runx2), osteopontin (Opn), and osteocalcin (Ocn) in vitro (1.5-2.5-fold increase compared with the control group in the most effective concentration group), which was consistent with the alkaline phosphatase (ALP) and alizarin red S (ARS) staining results. We implanted poly(sebacoyl diglyceride) (PSeD) combined with BD2 and rat bone tissue-derived mesenchymal stem cells (rBMSCs) under the back skin of rats and found that the inflammatory response was significantly lower with this combination than with the PSeD/rBMSCs scaffold without BD2 and the pure PSeD group and was similar to the control group. Importantly, when assessed in a critical-sized in vivo rat 8 m diameter calvaria defect model, a scaffold we developed combining bifunctional BD2 with porous organic polymer displayed an osteogenic effect that was 160-200% greater than the control group. The in vivo study results revealed a significant osteogenic response and antimicrobial effect and were consistent with the in vitro results. In summary, BD2 displayed a great potential of simultaneously promoting bone regeneration and preventing infection and could provide a viable alternative to traditional growth factors applied in bone defect repair.
Collapse
Affiliation(s)
- Yiyu Peng
- Department of Ophthalmology,
Ninth People’s Hospital, Shanghai
Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, P. R. China
| | - Lunhao Li
- Department of Ophthalmology,
Ninth People’s Hospital, Shanghai
Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, P. R. China
| | - Qingyue Yuan
- Department of Ophthalmology,
Ninth People’s Hospital, Shanghai
Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, P. R. China
| | - Ping Gu
- Department of Ophthalmology,
Ninth People’s Hospital, Shanghai
Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, P. R. China
| | - Zhengwei You
- State Key Laboratory for Modification of
Chemical Fibers and Polymer Materials, Shanghai Belt and Road Joint
Laboratory of Advanced Fiber and Low-dimension Materials (Donghua
University), College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China
| | - Ai Zhuang
- Department of Ophthalmology,
Ninth People’s Hospital, Shanghai
Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, P. R. China
- E-mail: . Tel: 18930843344. Fax: +8621-63134218 (A.Z.)
| | - Xiaoping Bi
- Department of Ophthalmology,
Ninth People’s Hospital, Shanghai
Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, P. R. China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, P. R. China
- E-mail: . Tel: +8621-63135606. Fax: +8621-63134218 (X.B.)
| |
Collapse
|
20
|
Kocijan R, Weigl M, Skalicky S, Geiger E, Ferguson J, Leinfellner G, Heimel P, Pietschmann P, Grillari J, Redl H, Hackl M. MicroRNA levels in bone and blood change during bisphosphonate and teriparatide therapy in an animal model of postmenopausal osteoporosis. Bone 2020; 131:115104. [PMID: 31683019 DOI: 10.1016/j.bone.2019.115104] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/27/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022]
Abstract
MicroRNAs control the activity of a variety of genes that are pivotal to bone metabolism. Therefore, the clinical utility of miRNAs as biomarkers and drug targets for bone diseases certainly merits further investigation. This study describes the use of an animal model of postmenopausal osteoporosis to generate a comprehensive dataset on miRNA regulation in bone tissue and peripheral blood during bone loss and specifically anti-resorptive and osteo-anabolic treatment. Forty-two Sprague-Dawley rats were randomized to SHAM surgery (n=10) or ovariectomy (OVX, n=32). Eight weeks after surgery, OVX animals were further randomized to anti-resorptive treatment with zoledronate (n=11), osteo-anabolic treatment with teriparatide (n=11), or vehicle treatment (n=10). After 12 weeks of treatment, bone and serum samples were used for microRNA analysis using next-generation sequencing (NGS), mRNA levels using RT-qPCR, and bone microarchitecture analysis using nanoCT. Ovariectomy resulted in loss of trabecular bone, which was fully rescued using osteo-anabolic treatment, and partially rescued using anti-resorptive treatment. NGS revealed that both, anti-resorptive and anabolic treatment had a significant impact on miRNA levels in bone tissue and serum: out of 426 detected miRNAs, 46 miRNAs were regulated by teriparatide treatment an d 10 by zoledronate treatment (p-adj.<0.1). Interestingly, teriparatide and zoledronate treatment were able to revert miRNA changes in tissue and serum of untreated OVX animals, such as the up-regulation of miR-203a-3p, a known osteo-inhibitory miRNA. We confirmed previously established mechanisms of miR-203a by analyzing its direct target Dlx5 in femoral head. Our data reveal a significant effect of ovariectomy-induced bone loss, as well as the two major types of anti-osteoporotic treatment on miRNA transcription in femoral head tissue. These changes are associated with altered activity of target genes relevant to bone formation, such as Dlx5. The observed effects of bone loss and treatment response on miRNA levels in bone are also reflected in the peripheral blood, suggesting the possibility of minimally-invasive monitoring of bone-derived miRNAs using liquid biopsies.
Collapse
Affiliation(s)
- Roland Kocijan
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria; Hanusch Hospital, 1st Medical Department, Heinrich Collin-Str. 30, 1140 Vienna, Austria
| | - Moritz Weigl
- TAmiRNA GmbH, Leberstrasse 20, 1110 Vienna, Austria
| | | | | | - James Ferguson
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Gabriele Leinfellner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Patrick Heimel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria; Karl Donath Laboratory for Hard Tissue and Biomaterial Research, Department of Oral Surgery, University Clinic of Dentistry, Austria
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria; Christian Doppler Laboratory on Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Donaueschingenstraße 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Matthias Hackl
- TAmiRNA GmbH, Leberstrasse 20, 1110 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria.
| |
Collapse
|
21
|
Shen X, Lei J, Du L. miR-31-5p may enhance the efficacy of chemotherapy with Taxol and cisplatin in TNBC. Exp Ther Med 2019; 19:375-383. [PMID: 31853314 DOI: 10.3892/etm.2019.8191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/19/2019] [Indexed: 12/14/2022] Open
Abstract
The limited efficacy of chemotherapy with Taxol (TAX) and cisplatin (DDP) in triple-negative breast cancer (TNBC) has prompted the investigation of combined therapies. Previous studies demonstrated that microRNA (miR)-31-5p is involved in various biological processes. In the present study, it was hypothesized that the overexpression of miR-31-5p may enhance the efficacy of chemotherapy. The expression levels of miR-31-5p in the TNBC cell lines MDA-MB-231 and MDA-MB-468 were measured using reverse transcription-quantitative PCR following transfection with miR-31-5p mimic or inhibitor. A Cell Counting Kit-8 and flow cytometry assays suggested that the overexpression of miR-31-5p inhibited cell proliferation and promoted apoptosis, and these effects were reversed by transfecting a miR-31-5p inhibitor into MDA-MB-231 and MDA-MB-468 cells. Furthermore, the overexpression of miR-31-5p increased the sensitivity of cells to chemotherapy, which exhibited an increase in apoptosis and in the expression level of Bax, and a decrease in the expression level of Bcl-2. Chemotherapy resistance induced by miR-31-5p inhibitor could be reversed by inhibiting the AKT signaling pathway in MDA-MB-231 and MDA-MB-468 cells. In conclusion, the present preclinical results indicated that targeting miR-31-5p may enhance the efficacy of TAX- and DDP-mediated chemotherapy in TNBC.
Collapse
Affiliation(s)
- Xiaowei Shen
- Department of General Surgery, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 200092, P.R. China
| | - Jiaqi Lei
- Department of General Surgery, Zhongshan Hospital Qingpu Branch, Fudan University, Shanghai 200092, P.R. China
| | - Lei Du
- Department of General Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
22
|
Bottani M, Banfi G, Lombardi G. Perspectives on miRNAs as Epigenetic Markers in Osteoporosis and Bone Fracture Risk: A Step Forward in Personalized Diagnosis. Front Genet 2019; 10:1044. [PMID: 31737038 PMCID: PMC6831724 DOI: 10.3389/fgene.2019.01044] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
Aging is associated with an increased incidence of age-related bone diseases. Current diagnostics (e.g., conventional radiology, biochemical markers), because limited in specificity and sensitivity, can distinguish between healthy or osteoporotic subjects but they are unable to discriminate among different underlying causes that lead to the same bone pathological condition (e.g., bone fracture risk). Among recent, more sensitive biomarkers, miRNAs — the non-coding RNAs involved in the epigenetic regulation of gene expression, have emerged as fundamental post-transcriptional modulators of bone development and homeostasis. Each identified miRNA carries out a specific role in osteoblast and osteoclast differentiation and functional pathways (osteomiRs). miRNAs bound to proteins or encapsulated in exosomes and/or microvesicles are released into the bloodstream and biological fluids where they can be detected and measured by highly sensitive and specific methods (e.g., quantitative PCR, next-generation sequencing). As such, miRNAs provide a prompt and easily accessible tool to determine the subject-specific epigenetic environment of a specific condition. Their use as biomarkers opens new frontiers in personalized medicine. While miRNAs circulating levels are lower than those found in the tissue/cell source, their quantification in biological fluids may be strategic in the diagnosis of diseases that affect tissues, such as bone, in which biopsy may be especially challenging. For a biomarker to be valuable in clinical practice and support medical decisions, it must be (easily) measurable, validated by independent studies, and strongly and significantly associated with a disease outcome. Currently, miRNAs analysis does not completely satisfy these criteria, however. Starting from in vitro and in vivo observations describing their biological role in bone cell development and metabolism, this review describes the potential use of bone-associated circulating miRNAs as biomarkers for determining predisposition, onset, and development of osteoporosis and bone fracture risk. Moreover, the review focuses on their clinical relevance and discusses the pre-analytical, analytical, and post-analytical issues in their measurement, which still limits their routine application. Taken together, research and clinical findings may be helpful for creating miRNA-based diagnostic tools in the diagnosis and treatment of bone diseases.
Collapse
Affiliation(s)
- Michela Bottani
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Moelcular Biology, Milano, Italy
| | - Giuseppe Banfi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Moelcular Biology, Milano, Italy.,Vita-Salute San Raffaele University, Milano, Italy
| | - Giovanni Lombardi
- IRCCS Istituto Ortopedico Galeazzi, Laboratory of Experimental Biochemistry & Moelcular Biology, Milano, Italy.,Department of Physiology & Pharmacology, Gdańsk University of Physical Education & Sport, Gdańsk, Poland
| |
Collapse
|
23
|
Hosseinpour S, He Y, Nanda A, Ye Q. MicroRNAs Involved in the Regulation of Angiogenesis in Bone Regeneration. Calcif Tissue Int 2019; 105:223-238. [PMID: 31175386 DOI: 10.1007/s00223-019-00571-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/01/2019] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) as a newly founded and thriving non-coding endogenous class of molecules which regulate many cellular pathways after transcription have been extensively investigated in regenerative medicine. In this systematic review, we sought to analyze miRNAs-mediated therapeutic approaches for influencing angiogenesis in bone tissue/bone regeneration. An electronic search in MEDLINE, Scopus, EMBASE, Cochrane library, web of science, and google scholar with no time limit were done on English publications. All types of original articles which a miRNA for angiogenesis in bone regeneration were included in our review. In the process of reviewing, we used PRISMA guideline and, SYRCLE's and science in risk assessment and policy tools for analyzing risk of bias. Among 751 initial retrieved records, 16 studies met the inclusion criteria and were fully assessed in this review. 275 miRNAs, one miRNA 195~497 cluster, and one Cysteine-rich 61 short hairpin RNA were differentially expressed during bone regeneration with 24 predicted targets reported in these studies. Among these miRNAs, miRNA-7b, -9, -21, -26a, -27a, -210, -378, -195~497 cluster, -378 and -675 positively promoted both angiogenesis and osteogenesis, whereas miRNA-10a, -222 and -494 inhibited both processes. The most common target was vasculoendothelial growth factor-signaling pathway. Recent evidence has demonstrated that miRNAs actively participated in angio-osteogenic coupling that can improve their therapeutic potentials for the treatment of bone-related diseases and bone regeneration. However, there is still need for further research to unravel the exact mechanisms.
Collapse
Affiliation(s)
- Sepanta Hosseinpour
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Yan He
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Ashwin Nanda
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia
| | - Qingsong Ye
- School of Dentistry, The University of Queensland, Herston, Brisbane, QLD, 4006, Australia.
| |
Collapse
|
24
|
Arriaga MA, Ding MH, Gutierrez AS, Chew SA. The Application of microRNAs in Biomaterial Scaffold-Based Therapies for Bone Tissue Engineering. Biotechnol J 2019; 14:e1900084. [PMID: 31166084 DOI: 10.1002/biot.201900084] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/28/2019] [Indexed: 12/13/2022]
Abstract
In recent years, the application of microRNAs (miRNAs) or anti-microRNAs (anti-miRNAs) that can induce expression of the runt-related transcription factor 2 (RUNX2), a master regulator of osteogenesis, has been investigated as a promising alternative bone tissue engineering strategy. In this review, biomaterial scaffold-based applications that have been used to deliver cells expressing miRNAs or anti-miRNAs that induce expression of RUNX2 for bone tissue engineering are discussed. An overview of the components of the scaffold-based therapies including the miRNAs/anti-miRNAs, cell types, gene delivery vectors, and scaffolds that have been applied are provided. To date, there have been nine miRNAs/anti-miRNAs (i.e., miRNA-26a, anti-miRNA-31, anti-miRNA-34a, miRNA-135, anti-miRNA-138, anti-miRNA-146a, miRNA-148b, anti-miRNA-221, and anti-miRNA-335) that have been incorporated into scaffold-based bone tissue engineering applications and investigated in an in vivo bone critical-sized defect model. For all of the biomaterial scaffold-based miRNA therapies that have been developed thus far, cells that are transfected or transduced with the miRNA/anti-miRNA are loaded into the scaffolds and implanted at the site of interest instead of locally delivering the miRNA/anti-miRNAs directly from the scaffolds. Thus, future work may focus on developing biomaterial scaffolds to deliver miRNAs or anti-miRNAs into cells in vivo.
Collapse
Affiliation(s)
- Marco A Arriaga
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - May-Hui Ding
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Astrid S Gutierrez
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| | - Sue Anne Chew
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, One West University Blvd, Brownsville, TX, 78520, USA
| |
Collapse
|
25
|
Vilariño-Feltrer G, Muñoz-Santa A, Conejero-García Á, Vallés-Lluch A. The effect of salt fusion processing variables on structural, physicochemical and biological properties of poly(glycerol sebacate) scaffolds. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2019.1636247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - Alba Muñoz-Santa
- Centre for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Álvaro Conejero-García
- Centre for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
| | - Ana Vallés-Lluch
- Centre for Biomaterials and Tissue Engineering, Universitat Politècnica de València, Valencia, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| |
Collapse
|
26
|
Wang J, Liu S, Li J, Zhao S, Yi Z. Roles for miRNAs in osteogenic differentiation of bone marrow mesenchymal stem cells. Stem Cell Res Ther 2019; 10:197. [PMID: 31253175 PMCID: PMC6599379 DOI: 10.1186/s13287-019-1309-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs), which were first discovered in bone marrow, are capable of differentiating into osteoblasts, chondrocytes, fat cells, and even myoblasts, and are considered multipotent cells. As a result of their potential for multipotential differentiation, self-renewal, immune regulation, and other effects, BMSCs have become an important source of seed cells for gene therapy, tissue engineering, cell replacement therapy, and regenerative medicine. MicroRNA (miRNA) is a highly conserved type of endogenous non-protein-encoding RNA of about 19-25 nucleotides in length, whose transcription process is independent of other genes. Generally, miRNA plays roles in regulating cell proliferation, differentiation, apoptosis, and development by binding to the 3' untranslated region of target mRNAs, whereby they can degrade or induce translational silencing. Although miRNAs play a regulatory role in various metabolic processes, they are not translated into proteins. Several studies have shown that miRNAs play an important role in the osteogenic differentiation of BMSCs. Herein, we describe in-depth studies of roles for miRNAs during the osteogenic differentiation of BMSCs, as they provide new theoretical and experimental rationales for bone tissue engineering and clinical treatment.
Collapse
Affiliation(s)
- Jicheng Wang
- Shaanxi Provincial People's Hospital, 256 Youyi West Road, Beilin, Xi'an, 710068, China.,Xi'an Medical University, Xi'an, 710068, China
| | - Shizhang Liu
- Shaanxi Provincial People's Hospital, 256 Youyi West Road, Beilin, Xi'an, 710068, China
| | - Jingyuan Li
- Shaanxi Provincial People's Hospital, 256 Youyi West Road, Beilin, Xi'an, 710068, China
| | - Song Zhao
- Shaanxi Provincial People's Hospital, 256 Youyi West Road, Beilin, Xi'an, 710068, China.,Xi'an Medical University, Xi'an, 710068, China
| | - Zhi Yi
- Shaanxi Provincial People's Hospital, 256 Youyi West Road, Beilin, Xi'an, 710068, China.
| |
Collapse
|
27
|
Shen H, Lu C, Shi J, Li H, Si J, Shen G. Satb2 expression in Foxc1-promoted osteogenic differentiation of MC3T3-E1 cells is negatively regulated by microRNA-103-3p. Acta Biochim Biophys Sin (Shanghai) 2019; 51:588-597. [PMID: 31089719 DOI: 10.1093/abbs/gmz037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Indexed: 12/12/2022] Open
Abstract
The forkhead transcription factor C1 (Foxc1) is a cell-fate-determining factor that controls cranial bone development and osteogenic differentiation. Previously, it was demonstrated that various microRNAs (miRNAs) play important roles in osteogenesis and regulate the complex process of osteogenic differentiation. However, it remains unclear how miRNA expression changes during Foxc1-promoted osteogenic differentiation. In this study, we successfully overexpressed the Foxc1 gene in MC3T3-E1 cells and investigated the alterations in the miRNA expression profile on day 3 after osteogenic induction by using a miRNA microarray. Nine downregulated miRNAs and eight upregulated miRNAs were found to be differentially expressed. Among these miRNAs, miR-103-3p was consistently downregulated in the Foxc1-overexpressing MC3T3-E1 cells and was identified as a negative regulator of osteogenic differentiation by using a gain- and lose-of-function assay. The special AT-rich sequence-binding protein 2 (Satb2), a pivotal osteogenic transcription factor, was identified as the miR-103-3p targeting gene and was verified by real-time polymerase chain reaction, western blot analysis, and luciferase assay. Overexpression of miR-103-3p markedly inhibited the expression of Satb2 and attenuated Foxc1-promoted osteogenic differentiation. Taken together, our results elucidated the miRNA expression profiles of MC3T3-E1 cells in the early stage of Foxc1-promoted osteogenic differentiation and suggested that miR-103-3p acts as a negative regulator of the osteogenic differentiation of MC3T3-E1 cells by directly targeting Satb2.
Collapse
Affiliation(s)
- Hongzhou Shen
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chenpei Lu
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jun Shi
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Hongliang Li
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiawen Si
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Guofang Shen
- Department of Oral and Craniomaxillofacial Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
28
|
Hadjiargyrou M, Komatsu DE. The Therapeutic Potential of MicroRNAs as Orthobiologics for Skeletal Fractures. J Bone Miner Res 2019; 34:797-809. [PMID: 30866092 PMCID: PMC6536331 DOI: 10.1002/jbmr.3708] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/04/2019] [Accepted: 02/23/2019] [Indexed: 12/19/2022]
Abstract
The repair of a fractured bone is critical to the well-being of humans. Failure of the repair process to proceed normally can lead to complicated fractures, exemplified by either a delay in union or a complete nonunion. Both of these conditions lead to pain, the possibility of additional surgery, and impairment of life quality. Additionally, work productivity decreases, income is reduced, and treatment costs increase, resulting in financial hardship. Thus, developing effective treatments for these difficult fractures or even accelerating the normal physiological repair process is warranted. Accumulating evidence shows that microRNAs (miRNAs), small noncoding RNAs, can serve as key regulatory molecules of fracture repair. In this review, a brief description of the fracture repair process and miRNA biogenesis is presented, as well as a summary of our current knowledge of the involvement of miRNAs in physiological fracture repair, osteoporotic fractures, and bone defect healing. Further, miRNA polymorphisms associated with fractures, miRNA presence in exosomes, and miRNAs as potential therapeutic orthobiologics are also discussed. This is a timely review as several miRNA-based therapeutics have recently entered clinical trials for nonskeletal applications and thus it is incumbent upon bone researchers to explore whether miRNAs can become the next class of orthobiologics for the treatment of skeletal fractures.
Collapse
Affiliation(s)
- Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY 11568-8000
| | - David E. Komatsu
- Department of Orthopaedics, Stony Brook University, Stony Brook, NY 11794-8181
| |
Collapse
|
29
|
Costa V, Carina V, Conigliaro A, Raimondi L, De Luca A, Bellavia D, Salamanna F, Setti S, Alessandro R, Fini M, Giavaresi G. miR-31-5p Is a LIPUS-Mechanosensitive MicroRNA that Targets HIF-1α Signaling and Cytoskeletal Proteins. Int J Mol Sci 2019; 20:E1569. [PMID: 30925808 PMCID: PMC6480017 DOI: 10.3390/ijms20071569] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 12/16/2022] Open
Abstract
The roles of low-intensity pulsed ultrasound (LIPUS) and microRNAs (miRNAs) on hMSCs commitments have already been investigated; however, the effects of the application of their co-treatments in an in vitro cell model are still unknown. Our previous studies demonstrated that (i) LIPUS modulated hMSCs cytoskeletal organization and (ii) miRNA-675-5p have a role in HIF-1α signaling modulation during hMSCs osteoblast commitment. We investigated for the first time the role of LIPUS as promoter tool for miRNA expression. Thanks to bioinformatic analysis, we identified miR-31-5p as a LIPUS-induced miRNA and investigated its role through in vitro studies of gain and loss of function. Results highlighted that LIPUS stimulation induced a hypoxia adaptive cell response, which determines a reorganization of cell membrane and cytoskeleton proteins. MiR-31-5p gain and loss of function studies, demonstrated as miR-31-5p overexpression, were able to induce hypoxic and cytoskeletal responses. Moreover, the co-treatments LIPUS and miR-31-5p inhibitor abolished the hypoxic responses including angiogenesis and the expression of Rho family proteins. MiR-31-5p was identified as a LIPUS-mechanosensitive miRNAs and may be considered a new therapeutic option to promote or abolish hypoxic response and cytoskeletal organization on hMSCs during the bone regeneration process.
Collapse
Affiliation(s)
- Viviana Costa
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Valeria Carina
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | - Alice Conigliaro
- Department of BioMedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), University of Palermo, 90100 Palermo, Italy.
| | | | - Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy.
| | | | - Francesca Salamanna
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, 40136 Bologna, Italy.
| | | | - Riccardo Alessandro
- Department of BioMedicine, Neuroscience and Advanced Diagnostics (Bi.N.D), University of Palermo, 90100 Palermo, Italy.
- Institute of Biomedicine and Molecular Immunology (IBIM), National Research Council, 90100 Palermo, Italy.
| | - Milena Fini
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, 40136 Bologna, Italy.
| | - Gianluca Giavaresi
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Preclinical and Surgical Studies, 40136 Bologna, Italy.
| |
Collapse
|
30
|
Liu H, Su H, Wang X, Hao W. MiR-148a regulates bone marrow mesenchymal stem cells-mediated fracture healing by targeting insulin-like growth factor 1. J Cell Biochem 2019; 120:1350-1361. [PMID: 30335895 DOI: 10.1002/jcb.27121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/04/2018] [Indexed: 01/24/2023]
Abstract
The purpose of this study was to investigate the underlying molecular mechanisms of fracture healing mediated by bone marrow mesenchymal stem cells. Differentially expressed microRNAs in acutely injured subjects and healthy volunteers were screened by microarray analysis. The dual luciferase reporter system was used to verify whether insulin-like growth factor 1 (IGF1) was the direct target gene regulated by miR-148a. The expression level of miR-148a and IGF1 after osteogenic differentiation was detected by quantitative real-time polymerase chain reaction. Western blot was used to determine the protein expression of bone markers, including IGF1, runt-related transcription factor 2 (Runx2), osteocalcin, and osteopontin in rat bone marrow-derived mesenchymal stem cells. Alkaline phosphatase and alizarin red staining was used to detect alkaline phosphatase activity and calcium deposition. An animal fracture model was used for in vivo experiments. MiR-148a was highly expressed in acutely injured subjects compared with healthy volunteers, and IGF1 was a target of miR-148a. Moreover, compared with the negative control group, IGF1 messenger RNA expression was significantly increased in the miR-148a antagomir group. During osteogenic differentiation, the expression of IGF1, Runx2, osteocalcin, and osteopontin was higher in the miR-148a antagomir group than other groups. In vivo experiments further confirmed that upregulation of IGF1 enhanced fracture healing efficiently by decreasing callus width and area and improving bone mineral density, maximum load, stiffness, and energy absorption. It was proved that IGF1 was the direct target gene of miR-148a, and the use of rat bone marrow-derived mesenchymal stem cells with low expression of miR-148a could improve fracture healing by upregulating IGF1.
Collapse
Affiliation(s)
- Hongzhi Liu
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Hao Su
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Xin Wang
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| | - Wei Hao
- Department of Orthopaedics and Traumatology, The Affiliated Yaitai Yuhuangding Hospital of Qingdao University Medical College, Yantai, Shandong, China
| |
Collapse
|
31
|
Sun X, Guo Q, Wei W, Robertson S, Yuan Y, Luo X. Current Progress on MicroRNA-Based Gene Delivery in the Treatment of Osteoporosis and Osteoporotic Fracture. Int J Endocrinol 2019; 2019:6782653. [PMID: 30962808 PMCID: PMC6431398 DOI: 10.1155/2019/6782653] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/28/2018] [Accepted: 12/31/2018] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence demonstrates that microRNAs, as important endogenous posttranscriptional regulators, are essential for bone remodeling and regeneration. Undoubtedly, microRNA-based gene therapies show great potential to become novel approaches against bone-related diseases, including osteoporosis and associated fractures. The major obstacles for continued advancement of microRNA-based therapies in clinical application include their poor in vivo stability, nonspecific biodistribution, and unwanted side effects. Appropriate chemical modifications and delivery vectors, which improve the biological performance and potency of microRNA-based drugs, hold the key to translating miRNA technologies into clinical practice. Thus, this review summarizes the current attempts and existing deficiencies of chemical modifications and delivery systems applied in microRNA-based therapies for osteoporosis and osteoporotic fractures to inform further explorations.
Collapse
Affiliation(s)
- Xi Sun
- Department of Endocrinology, The Third Xiangya Hospital of Central South University, 138# Tongzipo Road, Changsha, Hunan 410007, China
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| | - Wenhua Wei
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Stephen Robertson
- Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - Ying Yuan
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, 87# Xiangya Road, Changsha, Hunan 410008, China
| |
Collapse
|
32
|
Yu X, Liu S, Chen H, Zhao X, Chen X, Du Y, Li S. CGRP gene-modified rBMSCs show better osteogenic differentiation capacity in vitro. J Mol Histol 2018; 49:357-367. [PMID: 29846865 DOI: 10.1007/s10735-018-9775-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Calcitonin gene-related peptide (CGRP) is a marked and important neuropeptide expressed in nerve fibers during bone repair. This study investigated the role of CGRP overexpression on osteogenic differentiation of rat bone mesenchymal stem cells (rBMSCs). rBMSCs were infected with viral stocks of pLenO-DCE-CGRP (CGRP group) or pLenO-DCE (Vector group), while normal rBMSCs were used as a control. Transfection efficiency of rBMSCs was analyzed by flow cytometry. Cell proliferation was examined using a Cell Counting Kit-8 and flow cytometry. Expressions of alkaline phosphatase(ALP), bone sialoprotein (BSP) and Runt-related transcription factor 2(Runx2) in rBMSCs were detected at 1 and 2 weeks after mineral induction by real-time PCR and western blotting. Alizarin Red staining was applied at 28 days. The ratio of osteoprotegerin (OPG) to receptor activator of nuclear factor kappa B ligand (RANKL) was also detected to determine the underlying mechanism. pLenO-DCE-CGRP-induced rBMSCs stably overexpressing CGRP were successfully established. Overexpression of the CGRP gene significantly promoted rBMSC proliferation (p < 0.05). In addition, expressions of osteogenesis-related indexes were upregulated in the CGRP group (p < 0.05) compared with vector and control groups, and more mineralization nodules were observed in the CGRP group (p < 0.05). CGRP gene increased OPG and reduced RANKL in rBMSCs. Hence, the OPG/ RANKL ratio was increased in the CGRP group compared with the other two groups. CGRP gene-modified rBMSCs show better osteogenic differentiation capacity compared with rBMSCs in vitro.
Collapse
Affiliation(s)
- Xijiao Yu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Periodontology, School and Hospital of Stomatology, Shandong University, 44-1 West Wenhua Road, Jinan, 250012, Shandong, People's Republic of China.,Department of Endodontics, Jinan Stomatological Hospital, Jinan, Shandong, People's Republic of China
| | - Shuang Liu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Periodontology, School and Hospital of Stomatology, Shandong University, 44-1 West Wenhua Road, Jinan, 250012, Shandong, People's Republic of China
| | - Hui Chen
- Department of Endodontics, Jinan Stomatological Hospital, Jinan, Shandong, People's Republic of China
| | - Xinyu Zhao
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Periodontology, School and Hospital of Stomatology, Shandong University, 44-1 West Wenhua Road, Jinan, 250012, Shandong, People's Republic of China
| | - Xue Chen
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Periodontology, School and Hospital of Stomatology, Shandong University, 44-1 West Wenhua Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yi Du
- Department of Endodontics, Jinan Stomatological Hospital, Jinan, Shandong, People's Republic of China
| | - Shu Li
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Department of Periodontology, School and Hospital of Stomatology, Shandong University, 44-1 West Wenhua Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
33
|
Zheng CX, Sui BD, Hu CH, Qiu XY, Zhao P, Jin Y. Reconstruction of structure and function in tissue engineering of solid organs: Toward simulation of natural development based on decellularization. J Tissue Eng Regen Med 2018; 12:1432-1447. [PMID: 29701314 DOI: 10.1002/term.2676] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 10/13/2017] [Accepted: 04/16/2018] [Indexed: 12/21/2022]
Abstract
Failure of solid organs, such as the heart, liver, and kidney, remains a major cause of the world's mortality due to critical shortage of donor organs. Tissue engineering, which uses elements including cells, scaffolds, and growth factors to fabricate functional organs in vitro, is a promising strategy to mitigate the scarcity of transplantable organs. Within recent years, different construction strategies that guide the combination of tissue engineering elements have been applied in solid organ tissue engineering and have achieved much progress. Most attractively, construction strategy based on whole-organ decellularization has become a popular and promising approach, because the overall structure of extracellular matrix can be well preserved. However, despite the preservation of whole structure, the current constructs derived from decellularization-based strategy still perform partial functions of solid organs, due to several challenges, including preservation of functional extracellular matrix structure, implementation of functional recellularization, formation of functional vascular network, and realization of long-term functional integration. This review overviews the status quo of solid organ tissue engineering, including both advances and challenges. We have also put forward a few techniques with potential to solve the challenges, mainly focusing on decellularization-based construction strategy. We propose that the primary concept for constructing tissue-engineered solid organs is fabricating functional organs based on intact structure via simulating the natural development and regeneration processes.
Collapse
Affiliation(s)
- Chen-Xi Zheng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| | - Bing-Dong Sui
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Xin-Yu Qiu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| | - Pan Zhao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi, China.,Research and Development Center for Tissue Engineering, Fourth Military Medical University, Shaanxi, China
| |
Collapse
|
34
|
Li T, Liu ZL, Xiao M, Yang ZZ, Peng MZ, Li CD, Zhou XJ, Wang JW. Impact of bone marrow mesenchymal stem cell immunomodulation on the osteogenic effects of laponite. Stem Cell Res Ther 2018; 9:100. [PMID: 29642953 PMCID: PMC5896058 DOI: 10.1186/s13287-018-0818-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND With the development of osteoimmunology and bone tissue engineering (BTE), it has been recognized that the immunomodulatory properties of bone biomaterials have considerable impact in determining their fate after implantation. In this regard, the polarization of macrophages secondary to biomaterials is postulated to play a crucial role in modulating their osteogenesis; thus, strategies that may facilitate this process engender increasing levels of attention. Whereas a variety of reports highlight the immunomodulation of bone marrow mesenchymal stem cells (BMSCs) in cell therapy or their osteogenesis in BTE, few have focused on the effect of BMSCs in promoting osteogenesis in BTE through regulating the phenotype of macrophages. Accordingly, there is an urgent need to clarify the immunomodulatory properties of agents such as laponite (Lap), which is comprised of bioactive silicate nanoplatelets with excellent osteogenesis-inducing potential, to enhance their use in BTE. METHODS In the present study, we analyzed the osteoimmunomodulatory properties of Lap alone, as well as following the introduction of BMSCs into Lap, to determine whether BMSCs could modulate its immunomodulatory properties and promote osteogenesis. RESULTS It was found that the BMSCs reversed the polarization of murine-derived macrophage RAW 264.7 cells from M1 as induced by pure Lap to M2 and promoted osteogenesis. In vivo study confirmed that BMSCs combined with Lap initiated a less severe immune response and had an improved effect on bone regeneration compared with Lap alone, which corresponded with the in vitro evaluation. CONCLUSION These results suggest that BMSCs could ameliorate the inflammation induced by Lap and enhance its bone formation. The immunomodulatory characteristics of BMSCs suggest that these might be tailored as a new strategy to promote the osteogenic capacity of biomaterials.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Zhong Long Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Ming Xiao
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Ze Zheng Yang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Ming Zheng Peng
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Cui Di Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xiao Jun Zhou
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| | - Jin Wu Wang
- Shanghai Key Laboratory of Orthopaedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Room 701, No. 3 Building, 639 Zhizaoju Road, Shanghai, 200011 People’s Republic of China
| |
Collapse
|
35
|
Curtin CM, Castaño IM, O'Brien FJ. Scaffold-Based microRNA Therapies in Regenerative Medicine and Cancer. Adv Healthc Mater 2018; 7. [PMID: 29068566 DOI: 10.1002/adhm.201700695] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/21/2017] [Indexed: 12/17/2022]
Abstract
microRNA-based therapies are an advantageous strategy with applications in both regenerative medicine (RM) and cancer treatments. microRNAs (miRNAs) are an evolutionary conserved class of small RNA molecules that modulate up to one third of the human nonprotein coding genome. Thus, synthetic miRNA activators and inhibitors hold immense potential to finely balance gene expression and reestablish tissue health. Ongoing industry-sponsored clinical trials inspire a new miRNA therapeutics era, but progress largely relies on the development of safe and efficient delivery systems. The emerging application of biomaterial scaffolds for this purpose offers spatiotemporal control and circumvents biological and mechanical barriers that impede successful miRNA delivery. The nascent research in scaffold-mediated miRNA therapies translates know-how learnt from studies in antitumoral and genetic disorders as well as work on plasmid (p)DNA/siRNA delivery to expand the miRNA therapies arena. In this progress report, the state of the art methods of regulating miRNAs are reviewed. Relevant miRNA delivery vectors and scaffold systems applied to-date for RM and cancer treatment applications are discussed, as well as the challenges involved in their design. Overall, this progress report demonstrates the opportunity that exists for the application of miRNA-activated scaffolds in the future of RM and cancer treatments.
Collapse
Affiliation(s)
- Caroline M. Curtin
- Tissue Engineering Research Group; Department of Anatomy; Royal College of Surgeons in Ireland (RCSI); 123 St. Stephens Green Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin (TCD); Dublin 2 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; RCSI & TCD; Dublin 2 Ireland
| | - Irene Mencía Castaño
- Tissue Engineering Research Group; Department of Anatomy; Royal College of Surgeons in Ireland (RCSI); 123 St. Stephens Green Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin (TCD); Dublin 2 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; RCSI & TCD; Dublin 2 Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group; Department of Anatomy; Royal College of Surgeons in Ireland (RCSI); 123 St. Stephens Green Dublin 2 Ireland
- Trinity Centre for Bioengineering; Trinity College Dublin (TCD); Dublin 2 Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; RCSI & TCD; Dublin 2 Ireland
| |
Collapse
|
36
|
McAlinden A, Im GI. MicroRNAs in orthopaedic research: Disease associations, potential therapeutic applications, and perspectives. J Orthop Res 2018; 36:33-51. [PMID: 29194736 PMCID: PMC5840038 DOI: 10.1002/jor.23822] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that function to control many cellular processes by their ability to suppress expression of specific target genes. Tens to hundreds of target genes may be affected by one miRNA, thereby resulting in modulation of multiple pathways in any given cell type. Therefore, altered expression of miRNAs (i.e., during tissue development or in scenarios of disease or cellular stress) can have a profound impact on processes regulating cell differentiation, metabolism, proliferation, or apoptosis, for example. Over the past 5-10 years, thousands of reports have been published on miRNAs in cartilage and bone biology or disease, thus highlighting the significance of these non-coding RNAs in regulating skeletal development and homeostasis. For the purpose of this review, we will focus on miRNAs or miRNA families that have demonstrated function in vivo within the context of cartilage, bone or other orthopaedic-related tissues (excluding muscle). Specifically, we will discuss studies that have utilized miRNA transgenic mouse models or in vivo approaches to target a miRNA with the aim of altering conditions such as osteoarthritis, osteoporosis and bone fractures in rodents. We will not discuss miRNAs in the context skeletal cancers since this topic is worthy of a review of its own. Overall, we aim to provide a comprehensive description of where the field currently stands with respect to the therapeutic potential of specific miRNAs to treat orthopaedic conditions and current technologies to target and modify miRNA function in vivo. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:33-51, 2018.
Collapse
Affiliation(s)
- Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, Missouri 63110
| | - Gun-Il Im
- Department of Orthopaedic Surgery, Dongguk University Ilsan Hospital, 814 Siksa-Dong, Goyang, Korea
| |
Collapse
|
37
|
Yang Y, Fang S. Small non-coding RNAs-based bone regulation and targeting therapeutic strategies. Mol Cell Endocrinol 2017; 456:16-35. [PMID: 27888003 PMCID: PMC7116989 DOI: 10.1016/j.mce.2016.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/06/2016] [Accepted: 11/21/2016] [Indexed: 01/08/2023]
Abstract
Small non-coding RNAs, which are 20-25 nucleotide ribonucleic acids, have emerged as an important transformation in the biological evolution over almost three decades. microRNAs (miRNAs) and short interfering RNAs (siRNAs) are two significant categories of the small RNAs that exert important effects on bone endocrinology and skeletology. Therefore, clarifying the expression and function of these important molecules in bone endocrine physiology and pathology is of great significance for improving their potential therapeutic value for metabolism-associated bone diseases. In the present review, we highlight the recent advances made in understanding the function and molecular mechanism of these small non-coding RNAs in bone metabolism, especially their potentially therapeutic values in bone-related diseases.
Collapse
Affiliation(s)
- Ying Yang
- Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| | - Sijie Fang
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University, School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Huynh CT, Zheng Z, Nguyen MK, McMillan A, Yesilbag Tonga G, Rotello VM, Alsberg E. Cytocompatible Catalyst-Free Photodegradable Hydrogels for Light-Mediated RNA Release To Induce hMSC Osteogenesis. ACS Biomater Sci Eng 2017; 3:2011-2023. [DOI: 10.1021/acsbiomaterials.6b00796] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | | | | | | | - Gulen Yesilbag Tonga
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | - Vincent M. Rotello
- Department
of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, Massachusetts 01003, United States
| | | |
Collapse
|
39
|
Kerativitayanan P, Tatullo M, Khariton M, Joshi P, Perniconi B, Gaharwar AK. Nanoengineered Osteoinductive and Elastomeric Scaffolds for Bone Tissue Engineering. ACS Biomater Sci Eng 2017; 3:590-600. [DOI: 10.1021/acsbiomaterials.7b00029] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Marco Tatullo
- Maxillofacial
Unit, Calabrodental Clinic, 88900 Crotone, Italy
- Regenerative
Medicine Section, Tecnologica Research Institute, 88900 Crotone, Italy
| | | | | | | | - Akhilesh K. Gaharwar
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
40
|
Xie Q, Wei W, Ruan J, Ding Y, Zhuang A, Bi X, Sun H, Gu P, Wang Z, Fan X. Effects of miR-146a on the osteogenesis of adipose-derived mesenchymal stem cells and bone regeneration. Sci Rep 2017; 7:42840. [PMID: 28205638 PMCID: PMC5311870 DOI: 10.1038/srep42840] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/16/2017] [Indexed: 02/08/2023] Open
Abstract
Increasing evidence has indicated that bone morphogenetic protein 2 (BMP2) coordinates with microRNAs (miRNAs) to form intracellular networks regulating mesenchymal stem cells (MSCs) osteogenesis. This study aimed to identify specific miRNAs in rat adipose-derived mesenchymal stem cells (ADSCs) during BMP2-induced osteogenesis, we selected the most significantly down-regulated miRNA, miR-146a, to systematically investigate its role in regulating osteogenesis and bone regeneration. Overexpressing miR-146a notably repressed ADSC osteogenesis, whereas knocking down miR-146a greatly promoted this process. Drosophila mothers against decapentaplegic protein 4 (SMAD4), an important co-activator in the BMP signaling pathway, was miR-146a’s direct target and miR-146a exerted its repressive effect on SMAD4 through interacting with 3′-untranslated region (3′-UTR) of SMAD4 mRNA. Furthermore, knocking down SMAD4 attenuated the ability of miR-146a inhibitor to promote ADSC osteogenesis. Next, transduced ADSCs were incorporated with poly(sebacoyl diglyceride) (PSeD) porous scaffolds for repairing critical-sized cranial defect, the treatment of miR-146a inhibitor greatly enhanced ADSC-mediated bone regeneration with higher expression levels of SMAD4, Runt-related transcription factor 2 (Runx2) and Osterix in newly formed bone. In summary, our study showed that miR-146a negatively regulates the osteogenesis and bone regeneration from ADSCs both in vitro and in vivo.
Collapse
Affiliation(s)
- Qing Xie
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wei
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Ruan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Ding
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoping Bi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Sun
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Tsekoura EK, K C RB, Uludag H. Biomaterials to Facilitate Delivery of RNA Agents in Bone Regeneration and Repair. ACS Biomater Sci Eng 2016; 3:1195-1206. [PMID: 33440509 DOI: 10.1021/acsbiomaterials.6b00387] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bone healing after traumatic injuries or pathological diseases remains an important worldwide problem. In search of safer and more effective approaches to bone regeneration and repair, RNA-based therapeutic agents, specifically microRNAs (miRNAs) and short interfering RNA (siRNA), are beginning to be actively explored. In this review, we summarize current attempts to employ miRNAs and siRNAs in preclinical models of bone repair. We provide a summary of current limitations when attempting to utilize bioactive nucleic acids for therapeutic purposes and position the unique aspects of RNA reagents for clinical bone repair. Delivery strategies for RNA reagents are emphasized and nonviral carriers (biomaterial-based) employed to deliver such reagents are reviewed. Critical features of biomaterial carriers and various delivery technologies centered around nanoparticulate systems are highlighted. We conclude with the authors' perspectives on the future of the field, outlining main critical issues important to address as RNA reagents are explored for clinical applications.
Collapse
Affiliation(s)
- Eleni K Tsekoura
- Department of Chemical & Materials Engineering, Faculty of Engineering, ‡Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Remant Bahadur K C
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and §Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Hasan Uludag
- Department of Chemical & Materials Engineering, Faculty of Engineering, Department of Biomedical Engineering, Faculty of Medicine & Dentistry, and Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
42
|
Li KC, Lo SC, Sung LY, Liao YH, Chang YH, Hu YC. Improved calvarial bone repair by hASCs engineered with Cre/loxP-based baculovirus conferring prolonged BMP-2 and MiR-148b co-expression. J Tissue Eng Regen Med 2016; 11:3068-3077. [PMID: 27687795 DOI: 10.1002/term.2208] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/22/2016] [Accepted: 04/05/2016] [Indexed: 12/18/2022]
Abstract
Repairing large calvarial bone defects remains a challenging task. Previously, it was discovered that that miR-148b, when acting in concert with bone morphogenetic protein 2 (BMP-2), enhanced the osteogenesis of human adipose-derived stem cells (hASCs) and improved calvarial bone healing in nude mice. However, the molecular target of miR-148b remained elusive. Here it is revealed that miR-148b directly targets NOG, whose gene product (noggin) is an antagonist to BMPs and negatively regulates BMP-induced osteogenic differentiation and bone formation. A new Cre/loxP-based baculovirus system was employed to drive prolonged BMP-2 and miR-148b overexpression in hASCs, wherein the BMP-2 overexpression induced noggin expression but the concurrent miR-148b expression downregulated noggin, thus relieving the negative regulatory loop and ameliorating hASC osteogenesis without hindering hASC proliferation or triggering appreciable cytotoxicity. Implantation of the engineered hASCs coexpressing BMP-2 and miR-148b into nude mice enabled substantial repair of critical-size calvarial bone defects (4 mm diameter) at 12 weeks post-transplantation, filling 83% of the defect area, 75% of bone volume and restoring the bone density to 89% of the original bone density. Such superior healing effects indicate the potential of the Cre/loxP-based baculovirus-mediated BMP-2/miR-148b expression for calvarial bone repair. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kuei-Chang Li
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Chun Lo
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Yu Sung
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hsin Liao
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Han Chang
- Center for Tissue Engineering, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Orthopaedic, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yu-Chen Hu
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
43
|
Hackl M, Heilmeier U, Weilner S, Grillari J. Circulating microRNAs as novel biomarkers for bone diseases - Complex signatures for multifactorial diseases? Mol Cell Endocrinol 2016; 432:83-95. [PMID: 26525415 DOI: 10.1016/j.mce.2015.10.015] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/20/2015] [Accepted: 10/20/2015] [Indexed: 02/07/2023]
Abstract
Biomarkers are essential tools in clinical research and practice. Useful biomarkers must combine good measurability, validated association with biological processes or outcomes, and should support clinical decision making if used in clinical practice. Several types of validated biomarkers have been reported in the context of bone diseases. However, because these biomarkers face certain limitations there is an interest in the identification of novel biomarkers for bone diseases, specifically in those that are tightly linked to the disease pathology leading to increased fracture-risk. MicroRNAs (miRNAs) are the most abundant RNA species to be found in cell-free blood. Encapsulated within microvesicles or bound to proteins, circulating miRNAs are remarkably stable analytes that can be measured using gold-standard technologies such as quantitative polymerase-chain-reaction (qPCR). Nevertheless, the analysis of circulating miRNAs faces several pre-analytical as well as analytical challenges. From a biological view, there is accumulating evidence that miRNAs play essential roles in the regulation of various biological processes including bone homeostasis. Moreover, specific changes in miRNA transcription levels or miRNA secretory levels have been linked to the development and progression of certain bone diseases. Only recently, results from circulating miRNAs analysis in patients with osteopenia, osteoporosis and fragility fractures have been reported. By comparing these findings to studies on circulating miRNAs in cellular senescence and aging or muscle physiology and sarcopenia, several overlaps were observed. This suggests that signatures observed during osteoporosis might not be specific to the pathophysiology in bone, but rather integrate information from several tissue types. Despite these promising first data, more work remains to be done until circulating miRNAs can serve as established and robust diagnostic tools for bone diseases in clinical research, clinical routine and in personalized medicine.
Collapse
Affiliation(s)
| | - Ursula Heilmeier
- Musculoskeletal Quantitative Imaging Research Group, Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | | | - Johannes Grillari
- Evercyte GmbH, 1190 Vienna, Austria; Christian Doppler Laboratory for Biotechnology of Skin Aging, Department of Biotechnology, BOKU - University of Natural Resources and Life Sciences Vienna, 1190 Vienna, Austria.
| |
Collapse
|
44
|
Weilner S, Schraml E, Wieser M, Messner P, Schneider K, Wassermann K, Micutkova L, Fortschegger K, Maier AB, Westendorp R, Resch H, Wolbank S, Redl H, Jansen‐Dürr P, Pietschmann P, Grillari‐Voglauer R, Grillari J. Secreted microvesicular miR-31 inhibits osteogenic differentiation of mesenchymal stem cells. Aging Cell 2016; 15:744-54. [PMID: 27146333 PMCID: PMC4933673 DOI: 10.1111/acel.12484] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2016] [Indexed: 11/29/2022] Open
Abstract
Damage to cells and tissues is one of the driving forces of aging and age-related diseases. Various repair systems are in place to counteract this functional decline. In particular, the property of adult stem cells to self-renew and differentiate is essential for tissue homeostasis and regeneration. However, their functionality declines with age (Rando, 2006). One organ that is notably affected by the reduced differentiation capacity of stem cells with age is the skeleton. Here, we found that circulating microvesicles impact on the osteogenic differentiation capacity of mesenchymal stem cells in a donor-age-dependent way. While searching for factors mediating the inhibitory effect of elderly derived microvesicles on osteogenesis, we identified miR-31 as a crucial component. We demonstrated that miR-31 is present at elevated levels in the plasma of elderly and of osteoporosis patients. As a potential source of its secretion, we identified senescent endothelial cells, which are known to increase during aging in vivo (Erusalimsky, 2009). Endothelial miR-31 is secreted within senescent cell-derived microvesicles and taken up by mesenchymal stem cells where it inhibits osteogenic differentiation by knocking down its target Frizzled-3. Therefore, we suggest that microvesicular miR-31 in the plasma of elderly might play a role in the pathogenesis of age-related impaired bone formation and that miR-31 might be a valuable plasma-based biomarker for aging and for a systemic environment that does not favor cell-based therapies whenever osteogenesis is a limiting factor.
Collapse
Affiliation(s)
- Sylvia Weilner
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CenterDonaueschingenstrasse 13A‐1200ViennaAustria
- Evercyte GmbHMuthgasse 181190ViennaAustria
| | - Elisabeth Schraml
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
| | - Matthias Wieser
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
- ACIBMuthgasse 181190ViennaAustria
| | - Paul Messner
- Department of NanoBiotechnologyVienna Institute of BioTechnologyUniversity of Natural Resources and Life Sciences ViennaViennaAustria
| | - Karl Schneider
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CenterDonaueschingenstrasse 13A‐1200ViennaAustria
| | - Klemens Wassermann
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CenterDonaueschingenstrasse 13A‐1200ViennaAustria
| | - Lucia Micutkova
- Institute of Biomedical Aging ResearchAustrian Academy of SciencesViennaAustria
| | - Klaus Fortschegger
- Children's Cancer Research Institute (CCRI)St. Anna KinderkrebsforschungViennaAustria
| | - Andrea B. Maier
- Department of Medicine and Aged CareRoyal Melbourne HospitalUniversity of MelbourneMelbourneAustralia
- Department of Human Movement SciencesMOVE Research Institute AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Rudi Westendorp
- Department of public health and center for healthy aginguniversity of CopenhagenDenmark
| | - Heinrich Resch
- Department of Medicine 2St. Vincent Hospital1060ViennaAustria
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CenterDonaueschingenstrasse 13A‐1200ViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CenterDonaueschingenstrasse 13A‐1200ViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Pidder Jansen‐Dürr
- Institute of Biomedical Aging ResearchAustrian Academy of SciencesViennaAustria
| | - Peter Pietschmann
- Department of Pathophysiology and Allergy ResearchCenter of PathophysiologyInfectiology and ImmunologyMedical University of Vienna1090ViennaAustria
| | - Regina Grillari‐Voglauer
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
- Evercyte GmbHMuthgasse 181190ViennaAustria
- ACIBMuthgasse 181190ViennaAustria
| | - Johannes Grillari
- Department of BiotechnologyBOKU ‐ University of Natural Resources and Life Sciences ViennaMuthgasse 181190ViennaAustria
- Evercyte GmbHMuthgasse 181190ViennaAustria
- Austrian Cluster for Tissue RegenerationViennaAustria
| |
Collapse
|
45
|
STEPICHEVA NADEZDAA, SONG JIAL. Function and regulation of microRNA-31 in development and disease. Mol Reprod Dev 2016; 83:654-74. [PMID: 27405090 PMCID: PMC6040227 DOI: 10.1002/mrd.22678] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs that orchestrate numerous cellular processes both under normal physiological conditions as well as in diseases. This review summarizes the functional roles and transcriptional regulation of the highly evolutionarily conserved miRNA, microRNA-31 (miR-31). miR-31 is an important regulator of embryonic implantation, development, bone and muscle homeostasis, and immune system function. Its own regulation is disrupted during the onset and progression of cancer and autoimmune disorders such as psoriasis and systemic lupus erythematosus. Limited studies suggest that miR-31 is transcriptionally regulated by epigenetics, such as methylation and acetylation, as well as by a number of transcription factors. Overall, miR-31 regulates diverse cellular and developmental processes by targeting genes involved in cell proliferation, apoptosis, cell differentiation, and cell motility. Mol. Reprod. Dev. 83: 654-674, 2016 © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - JIA L. SONG
- Department of Biological Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
46
|
Mencía Castaño I, Curtin CM, Duffy GP, O'Brien FJ. Next generation bone tissue engineering: non-viral miR-133a inhibition using collagen-nanohydroxyapatite scaffolds rapidly enhances osteogenesis. Sci Rep 2016; 6:27941. [PMID: 27297802 PMCID: PMC4906381 DOI: 10.1038/srep27941] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 05/26/2016] [Indexed: 01/08/2023] Open
Abstract
Bone grafts are the second most transplanted materials worldwide at a global cost to healthcare systems valued over $30 billion every year. The influence of microRNAs in the regenerative capacity of stem cells offers vast therapeutic potential towards bone grafting; however their efficient delivery to the target site remains a major challenge. This study describes how the functionalisation of porous collagen-nanohydroxyapatite (nHA) scaffolds with miR-133a inhibiting complexes, delivered using non-viral nHA particles, enhanced human mesenchymal stem cell-mediated osteogenesis through the novel focus on a key activator of osteogenesis, Runx2. This study showed enhanced Runx2 and osteocalcin expression, as well as increased alkaline phosphatase activity and calcium deposition, thus demonstrating a further enhanced therapeutic potential of a biomaterial previously optimised for bone repair applications. The promising features of this platform offer potential for a myriad of applications beyond bone repair and tissue engineering, thus presenting a new paradigm for microRNA-based therapeutics.
Collapse
Affiliation(s)
- Irene Mencía Castaño
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI &TCD, Dublin 2, Ireland
| | - Caroline M Curtin
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI &TCD, Dublin 2, Ireland
| | - Garry P Duffy
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI &TCD, Dublin 2, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephens Green, Dublin 2, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin (TCD), College Green, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI &TCD, Dublin 2, Ireland
| |
Collapse
|
47
|
Huynh CT, Nguyen MK, Naris M, Tonga GY, Rotello VM, Alsberg E. Light-triggered RNA release and induction of hMSC osteogenesis via photodegradable, dual-crosslinked hydrogels. Nanomedicine (Lond) 2016; 11:1535-50. [PMID: 27246686 PMCID: PMC5827787 DOI: 10.2217/nnm-2016-0088] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 05/04/2016] [Indexed: 12/30/2022] Open
Abstract
AIM To engineer a photodegradable hydrogel system for actively controlled release of bioactive unmodified RNA at designated time points to induce hMSC osteogenesis. MATERIALS & METHODS RNA/polyethylenimine complexes were loaded into dual-crosslinked photodegradable hydrogels to examine the capacity of UV light application to trigger their release. The ability of released RNA to drive hMSC osteogenic differentiation was also investigated. RESULTS & CONCLUSION RNA release from photodegradable hydrogels was accelerated upon UV application, which was not observed in non-photodegradable hydrogels. Regardless of the presence of UV light, released siGFP exhibited high bioactivity by silencing GFP expression in HeLa cells. Importantly, siNoggin or miRNA-20a released from the hydrogels induced hMSC osteogenesis. This system provides a potentially valuable physician/patient-controlled 'on-demand' RNA delivery platform for biomedical applications.
Collapse
Affiliation(s)
- Cong Truc Huynh
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Minh Khanh Nguyen
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Mantas Naris
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Gulen Yesilbag Tonga
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Vincent M Rotello
- Department of Orthopaedic Surgery, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|
48
|
Wang Z, Lin M, Xie Q, Sun H, Huang Y, Zhang D, Yu Z, Bi X, Chen J, Wang J, Shi W, Gu P, Fan X. Electrospun silk fibroin/poly(lactide-co-ε-caprolactone) nanofibrous scaffolds for bone regeneration. Int J Nanomedicine 2016; 11:1483-500. [PMID: 27114708 PMCID: PMC4833379 DOI: 10.2147/ijn.s97445] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Tissue engineering has become a promising therapeutic approach for bone regeneration. Nanofibrous scaffolds have attracted great interest mainly due to their structural similarity to natural extracellular matrix (ECM). Poly(lactide-co-ε-caprolactone) (PLCL) has been successfully used in bone regeneration, but PLCL polymers are inert and lack natural cell recognition sites, and the surface of PLCL scaffold is hydrophobic. Silk fibroin (SF) is a kind of natural polymer with inherent bioactivity, and supports mesenchymal stem cell attachment, osteogenesis, and ECM deposition. Therefore, we fabricated hybrid nanofibrous scaffolds by adding different weight ratios of SF to PLCL in order to find a scaffold with improved properties for bone regeneration. Methods Hybrid nanofibrous scaffolds were fabricated by blending different weight ratios of SF with PLCL. Human adipose-derived stem cells (hADSCs) were seeded on SF/PLCL nanofibrous scaffolds of various ratios for a systematic evaluation of cell adhesion, proliferation, cytotoxicity, and osteogenic differentiation; the efficacy of the composite of hADSCs and scaffolds in repairing critical-sized calvarial defects in rats was investigated. Results The SF/PLCL (50/50) scaffold exhibited favorable tensile strength, surface roughness, and hydrophilicity, which facilitated cell adhesion and proliferation. Moreover, the SF/PLCL (50/50) scaffold promoted the osteogenic differentiation of hADSCs by elevating the expression levels of osteogenic marker genes such as BSP, Ocn, Col1A1, and OPN and enhanced ECM mineralization. In vivo assays showed that SF/PLCL (50/50) scaffold improved the repair of the critical-sized calvarial defect in rats, resulting in increased bone volume, higher trabecular number, enhanced bone mineral density, and increased new bone areas, compared with the pure PLCL scaffold. Conclusion The SF/PLCL (50/50) nanofibrous scaffold facilitated hADSC proliferation and osteogenic differentiation in vitro and further promoted new bone formation in vivo, suggesting that the SF/PLCL (50/50) nanofibrous scaffold holds great potential in bone tissue regeneration.
Collapse
Affiliation(s)
- Zi Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Ming Lin
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Qing Xie
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Hao Sun
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Yazhuo Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - DanDan Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Zhang Yu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Xiaoping Bi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Junzhao Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Jing Wang
- Biomaterials and Tissue Engineering Laboratory, College of Chemistry & Chemical Engineering and Biotechnology, Donghua University, Shanghai, People's Republic of China
| | - Wodong Shi
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Ping Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Donghua University, Shanghai, People's Republic of China
| |
Collapse
|
49
|
Kim YD, Pofali P, Park TE, Singh B, Cho K, Maharjan S, Dandekar P, Jain R, Choi YJ, Arote R, Cho CS. Gene therapy for bone tissue engineering. Tissue Eng Regen Med 2016; 13:111-125. [PMID: 30603391 PMCID: PMC6170855 DOI: 10.1007/s13770-016-9063-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/24/2015] [Accepted: 09/29/2015] [Indexed: 02/06/2023] Open
Abstract
Gene therapy holds a great promise and has been extensively investigated to improve bone formation and regeneration therapies in bone tissue engineering. A variety of osteogenic genes can be delivered by combining different vectors (viral or non-viral), scaffolds and delivery methodologies. Ex vivo & in vivo gene enhanced tissue engineering approaches have led to successful osteogenic differentiation and bone formation. In this article, we review recent advances of gene therapy-based bone tissue engineering discussing strengths and weaknesses of various strategies as well as general overview of gene therapy.
Collapse
Affiliation(s)
- Young-Dong Kim
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Korea
| | - Prasad Pofali
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Tae-Eun Park
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Kihyun Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Sushila Maharjan
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Rohidas Arote
- Department of Molecular Genetics, School of Dentistry, Seoul National University, Seoul, Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
50
|
Nakasa T, Yoshizuka M, Andry Usman M, Elbadry Mahmoud E, Ochi M. MicroRNAs and Bone Regeneration. Curr Genomics 2016; 16:441-52. [PMID: 27019619 PMCID: PMC4765532 DOI: 10.2174/1389202916666150817213630] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/18/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022] Open
Abstract
Bone has multiple functions, both morphologically and physiologically, and it frequently features in the pathological condition, including fracture and osteoporosis. For bone regeneration therapy, the regulation of osteoblast differentiation is important. MicroRNA (miRNA)s are short noncoding RNA which regulate gene expression at the post-transcriptional level. MiRNAs play an important role not only in a variety of other cellular processes including differentiation, proliferation, and apoptosis but also in the pathogenesis of human diseases. Recently, miRNAs have been known to participate in osteoblast differentiation by regulating several signaling pathways including transcription
factors. New insight into the mechanism during osteogenes is affected by miRNAs has been gained. Moreover, therapeutic trials for bone diseases including osteoporosis, fracture and bone defects targeting miRNAs have been examined in animal models. MiRNA therapy will enable development of a bone regeneration therapy.
Collapse
Affiliation(s)
- Tomoyuki Nakasa
- Department of Orthopaedics Surgery, Integrated Health Sciences, Institute of Biomedical & health Science, Hiroshima University, 1-2-3 Kasumi Minami-ku, Hiroshima City, 734-8551, Japan
| | - Masaaki Yoshizuka
- Department of Orthopaedics Surgery, Integrated Health Sciences, Institute of Biomedical & health Science, Hiroshima University, 1-2-3 Kasumi Minami-ku, Hiroshima City, 734-8551, Japan
| | - Muhammad Andry Usman
- Department of Orthopaedics Surgery, Integrated Health Sciences, Institute of Biomedical & health Science, Hiroshima University, 1-2-3 Kasumi Minami-ku, Hiroshima City, 734-8551, Japan
| | - Elhussein Elbadry Mahmoud
- Department of Orthopaedics Surgery, Integrated Health Sciences, Institute of Biomedical & health Science, Hiroshima University, 1-2-3 Kasumi Minami-ku, Hiroshima City, 734-8551, Japan
| | - Mitsuo Ochi
- Department of Orthopaedics Surgery, Integrated Health Sciences, Institute of Biomedical & health Science, Hiroshima University, 1-2-3 Kasumi Minami-ku, Hiroshima City, 734-8551, Japan
| |
Collapse
|