1
|
Zhang D, Jin X, Ma X, Qiu Y, Ma W, Dai X, Zhang Z. Tumour necrosis factor α regulates the miR-27a-3p-Sfrp1 axis in a mouse model of osteoporosis. Exp Physiol 2024; 109:1109-1123. [PMID: 38748896 PMCID: PMC11215474 DOI: 10.1113/ep090311] [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: 01/05/2022] [Accepted: 03/01/2024] [Indexed: 07/02/2024]
Abstract
Osteoporosis is a metabolic bone disease that involves gradual loss of bone density and mass, thus resulting in increased fragility and risk of fracture. Inflammatory cytokines, such as tumour necrosis factor α (TNF-α), inhibit osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), and several microRNAs are implicated in osteoporosis development. This study aimed to explore the correlation between TNF-α treatment and miR-27a-3p expression in BMSC osteogenesis and further understand their roles in osteoporosis. An osteoporosis animal model was established using ovariectomized (OVX) mice. Compared with Sham mice, the OVX mice had a significantly elevated level of serum TNF-α and decreased level of bone miR-27a-3p, and in vitro TNF-α treatment inhibited miR-27a-3p expression in BMSCs. In addition, miR-27a-3p promoted osteogenic differentiation of mouse BMSCs in vitro, as evidenced by alkaline phosphatase staining and Alizarin Red-S staining, as well as enhanced expression of the osteogenic markers Runx2 and Osterix. Subsequent bioinformatics analysis combined with experimental validation identified secreted frizzled-related protein 1 (Sfrp1) as a downstream target of miR-27a-3p. Sfrp1 overexpression significantly inhibited the osteogenic differentiation of BMSCs in vitro and additional TNF-α treatment augmented this inhibition. Moreover, Sfrp1 overexpression abrogated the promotive effect of miR-27a-3p on the osteogenic differentiation of BMSCs. Furthermore, the miR-27a-3p-Sfrp1 axis was found to exert its regulatory function in BMSC osteogenic differentiation via regulating Wnt3a-β-catenin signalling. In summary, this study revealed that TNF-α regulated a novel miR-27a-3p-Sfrp1 axis in osteogenic differentiation of BMSCs. The data provide new insights into the development of novel therapeutic strategies for osteoporosis.
Collapse
Affiliation(s)
- Dang‐Feng Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Xiao‐Na Jin
- Department of NursingXi'an International UniversityXi'anShaanxiChina
| | - Xing Ma
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Yu‐Sheng Qiu
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Wei Ma
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Xing Dai
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Zhi Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
2
|
Hussain MS, Shaikh NK, Agrawal M, Tufail M, Bisht AS, Khurana N, Kumar R. Osteomyelitis and non-coding RNAS: A new dimension in disease understanding. Pathol Res Pract 2024; 255:155186. [PMID: 38350169 DOI: 10.1016/j.prp.2024.155186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/27/2024] [Accepted: 01/31/2024] [Indexed: 02/15/2024]
Abstract
Osteomyelitis, a debilitating bone infection, presents considerable clinical challenges due to its intricate etiology and limited treatment options. Despite strides in surgical and chemotherapeutic interventions, the treatment landscape for osteomyelitis remains unsatisfactory. Recent attention has focused on the role of non-coding RNAs (ncRNAs) in the pathogenesis and progression of osteomyelitis. This review consolidates current knowledge on the involvement of distinct classes of ncRNAs, including microRNAs, long ncRNAs, and circular RNAs, in the context of osteomyelitis. Emerging evidence from various studies underscores the potential of ncRNAs in orchestrating gene expression and influencing the differentiation of osteoblasts and osteoclasts, pivotal processes in bone formation. The review initiates by elucidating the regulatory functions of ncRNAs in fundamental cellular processes such as inflammation, immune response, and bone remodeling, pivotal in osteomyelitis pathology. It delves into the intricate network of interactions between ncRNAs and their target genes, illuminating how dysregulation contributes to the establishment and persistence of osteomyelitic infections. Understanding their regulatory roles may pave the way for targeted diagnostic tools and innovative therapeutic interventions, promising a paradigm shift in the clinical approach to this challenging condition. Additionally, we delve into the promising therapeutic applications of these molecules, envisioning novel diagnostic and treatment approaches to enhance the management of this challenging bone infection.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Department of Pharmacology, School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan 302017, India
| | - Nusrat K Shaikh
- Department of Quality Assurance, Smt. N. M. Padalia Pharmacy College, Ahmedabad, 382210 Gujarat, India
| | - Mohit Agrawal
- Department of Pharmacology, School of Medical & Allied Sciences, K.R. Mangalam University, Gurugram 122103, India
| | - Muhammad Tufail
- Department of Oral and Maxillofacial Surgery, Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China.
| | - Ajay Singh Bisht
- School of Pharmaceutical Sciences, Shri Guru Ram Rai University, Patel Nagar, Dehradun, Uttarakhand 248001, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
3
|
Kechagias S, Theodoridis K, Broomfield J, Malpartida-Cardenas K, Reid R, Georgiou P, van Arkel RJ, Jeffers JRT. The effect of nodal connectivity and strut density within stochastic titanium scaffolds on osteogenesis. Front Bioeng Biotechnol 2023; 11:1305936. [PMID: 38107615 PMCID: PMC10721980 DOI: 10.3389/fbioe.2023.1305936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023] Open
Abstract
Modern orthopaedic implants use lattice structures that act as 3D scaffolds to enhance bone growth into and around implants. Stochastic scaffolds are of particular interest as they mimic the architecture of trabecular bone and can combine isotropic properties and adjustable structure. The existing research mainly concentrates on controlling the mechanical and biological performance of periodic lattices by adjusting pore size and shape. Still, less is known on how we can control the performance of stochastic lattices through their design parameters: nodal connectivity, strut density and strut thickness. To elucidate this, four lattice structures were evaluated with varied strut densities and connectivity, hence different local geometry and mechanical properties: low apparent modulus, high apparent modulus, and two with near-identical modulus. Pre-osteoblast murine cells were seeded on scaffolds and cultured in vitro for 28 days. Cell adhesion, proliferation and differentiation were evaluated. Additionally, the expression levels of key osteogenic biomarkers were used to assess the effect of each design parameter on the quality of newly formed tissue. The main finding was that increasing connectivity increased the rate of osteoblast maturation, tissue formation and mineralisation. In detail, doubling the connectivity, over fixed strut density, increased collagen type-I by 140%, increased osteopontin by 130% and osteocalcin by 110%. This was attributed to the increased number of acute angles formed by the numerous connected struts, which facilitated the organization of cells and accelerated the cell cycle. Overall, increasing connectivity and adjusting strut density is a novel technique to design stochastic structures which combine a broad range of biomimetic properties and rapid ossification.
Collapse
Affiliation(s)
- Stylianos Kechagias
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | | | - Joseph Broomfield
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Kenny Malpartida-Cardenas
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Ruth Reid
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Pantelis Georgiou
- Centre for Bio Inspired Technology, Department of Electrical and Electronic Engineering, Imperial College London, London, United Kingdom
| | - Richard J. van Arkel
- Department of Mechanical Engineering, Imperial College London, London, United Kingdom
| | | |
Collapse
|
4
|
Botor M, Auguściak-Duma A, Lesiak M, Sieroń Ł, Dziedzic-Kowalska A, Witecka J, Asman M, Madetko-Talowska A, Bik-Multanowski M, Galicka A, Sieroń AL, Gawron K. Analysis of miRNAs in Osteogenesis imperfecta Caused by Mutations in COL1A1 and COL1A2: Insights into Molecular Mechanisms and Potential Therapeutic Targets. Pharmaceuticals (Basel) 2023; 16:1414. [PMID: 37895885 PMCID: PMC10609877 DOI: 10.3390/ph16101414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023] Open
Abstract
Osteogenesis imperfecta (OI) is a group of connective tissue disorders leading to abnormal bone formation, mainly due to mutations in genes encoding collagen type I (Col I). Osteogenesis is regulated by a number of molecules, including microRNAs (miRNAs), indicating their potential as targets for OI therapy. The goal of this study was to identify and analyze the expression profiles of miRNAs involved in bone extracellular matrix (ECM) regulation in patients diagnosed with OI type I caused by mutations in COL1A1 or COL1A2. Primary skin fibroblast cultures were used for DNA purification and sequence analysis, followed by analysis of miRNA expression. Sequencing analysis revealed mutations of the COL1A1 or COL1A2 genes in all OI patients, including four previously unreported. Amongst the 40 miRNAs analyzed, 9 were identified exclusively in OI cells and 26 in both OI patients and the controls. In the latter case, the expression of six miRNAs (hsa-miR-10b-5p, hsa-miR-19a-3p, hsa-miR-19b-3p, has-miR-204-5p, has-miR-216a-5p, and hsa-miR-449a) increased, while four (hsa-miR-129-5p, hsa-miR-199b-5p, hsa-miR-664a-5p, and hsa-miR-30a-5p) decreased significantly in OI cells in comparison to their expression in the control cells. The identified mutations and miRNA expression profiles shed light on the intricate processes governing bone formation and ECM regulation, paving the way for further research and potential therapeutic advancements in OI and other genetic diseases related to bone abnormality management.
Collapse
Affiliation(s)
- Malwina Botor
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| | - Aleksandra Auguściak-Duma
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| | - Marta Lesiak
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| | - Łukasz Sieroń
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| | - Agata Dziedzic-Kowalska
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| | - Joanna Witecka
- Department of Parasitology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 41-200 Sosnowiec, Poland;
| | - Marek Asman
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Zabrze, Poland;
| | - Anna Madetko-Talowska
- Department of Medical Genetics, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.-T.); (M.B.-M.)
| | - Mirosław Bik-Multanowski
- Department of Medical Genetics, Jagiellonian University Medical College, 30-663 Krakow, Poland; (A.M.-T.); (M.B.-M.)
| | - Anna Galicka
- Department of Medical Chemistry, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | - Aleksander L. Sieroń
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| | - Katarzyna Gawron
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland; (A.A.-D.); (M.L.); (Ł.S.); (A.L.S.)
| |
Collapse
|
5
|
Philippe S, Delay M, Macian N, Morel V, Pickering ME. Common miRNAs of Osteoporosis and Fibromyalgia: A Review. Int J Mol Sci 2023; 24:13513. [PMID: 37686318 PMCID: PMC10488272 DOI: 10.3390/ijms241713513] [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: 07/10/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
A significant clinical association between osteoporosis (OP) and fibromyalgia (FM) has been shown in the literature. Given the need for specific biomarkers to improve OP and FM management, common miRNAs might provide promising tracks for future prevention and treatment. The aim of this review is to identify miRNAs described in OP and FM, and dysregulated in the same direction in both pathologies. The PubMed database was searched until June 2023, with a clear mention of OP, FM, and miRNA expression. Clinical trials, case-control, and cross-sectional studies were included. Gray literature was not searched. Out of the 184 miRNAs found in our research, 23 are shared by OP and FM: 7 common miRNAs are dysregulated in the same direction for both pathologies (3 up-, 4 downregulated). The majority of these common miRNAs are involved in the Wnt pathway and the cholinergic system and a possible link has been highlighted. Further studies are needed to explore this relationship. Moreover, the harmonization of technical methods is necessary to confirm miRNAs shared between OP and FM.
Collapse
Affiliation(s)
- Soline Philippe
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
| | - Marine Delay
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
- Inserm 1107, Neuro-Dol, University Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Nicolas Macian
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
| | - Véronique Morel
- Platform of Clinical Investigation Department, Inserm CIC 1405, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France; (S.P.); (M.D.); (N.M.); (V.M.)
| | - Marie-Eva Pickering
- Rheumatology Department, University Hospital Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| |
Collapse
|
6
|
Huber J, Longaker MT, Quarto N. Circulating and extracellular vesicle-derived microRNAs as biomarkers in bone-related diseases. Front Endocrinol (Lausanne) 2023; 14:1168898. [PMID: 37293498 PMCID: PMC10244776 DOI: 10.3389/fendo.2023.1168898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/31/2023] [Indexed: 06/10/2023] Open
Abstract
MicroRNAs (miRNA) are small non-coding RNA molecules that regulate posttranscriptional gene expression by repressing messengerRNA-targets. MiRNAs are abundant in many cell types and are secreted into extracellular fluids, protected from degradation by packaging in extracellular vesicles. These circulating miRNAs are easily accessible, disease-specific and sensitive to small changes, which makes them ideal biomarkers for diagnostic, prognostic, predictive or monitoring purposes. Specific miRNA signatures can be reflective of disease status and development or indicators of poor treatment response. This is especially important in malignant diseases, as the ease of accessibility of circulating miRNAs circumvents the need for invasive tissue biopsy. In osteogenesis, miRNAs can act either osteo-enhancing or osteo-repressing by targeting key transcription factors and signaling pathways. This review highlights the role of circulating and extracellular vesicle-derived miRNAs as biomarkers in bone-related diseases, with a specific focus on osteoporosis and osteosarcoma. To this end, a comprehensive literature search has been performed. The first part of the review discusses the history and biology of miRNAs, followed by a description of different types of biomarkers and an update of the current knowledge of miRNAs as biomarkers in bone related diseases. Finally, limitations of miRNAs biomarker research and future perspectives will be presented.
Collapse
Affiliation(s)
- Julika Huber
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Plastic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Natalina Quarto
- Hagey Laboratory for Pediatric Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
7
|
Lai G, Zhao R, Zhuang W, Hou Z, Yang Z, He P, Wu J, Sang H. BMSC-derived exosomal miR-27a-3p and miR-196b-5p regulate bone remodeling in ovariectomized rats. PeerJ 2022; 10:e13744. [PMID: 36168439 PMCID: PMC9509671 DOI: 10.7717/peerj.13744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/27/2022] [Indexed: 01/22/2023] Open
Abstract
Background In the bone marrow microenvironment of postmenopausal osteoporosis (PMOP), bone marrow mesenchymal stem cell (BMSC)-derived exosomal miRNAs play an important role in bone formation and bone resorption, although the pathogenesis has yet to be clarified. Methods BMSC-derived exosomes from ovariectomized rats (OVX-Exo) and sham-operated rats (Sham-Exo) were co-cultured with bone marrow-derived macrophages to study their effects on osteoclast differentiation. Next-generation sequencing was utilized to identify the differentially expressed miRNAs (DE-miRNAs) between OVX-Exo and Sham-Exo, while target genes were analyzed using bioinformatics. The regulatory effects of miR-27a-3p and miR-196b-5p on osteogenic differentiation of BMSCs and osteoclast differentiation were verified by gain-of-function and loss-of-function analyses. Results Osteoclast differentiation was significantly enhanced in the OVX-Exo treatment group compared to the Sham-Exo group. Twenty DE-miRNAs were identified between OVX-Exo and Sham-Exo, among which miR-27a-3p and miR-196b-5p promoted the expressions of osteogenic differentiation markers in BMSCs. In contrast, knockdown of miR-27a-3p and miR-196b-5p increased the expressions of osteoclastic markers in osteoclast. These 20 DE-miRNAs were found to target 11435 mRNAs. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that these target genes were involved in several biological processes and osteoporosis-related signaling pathways. Conclusion BMSC-derived exosomal miR-27a-3p and miR-196b-5p may play a positive regulatory role in bone remodeling.
Collapse
Affiliation(s)
- Guohua Lai
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Renli Zhao
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weida Zhuang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zuoxu Hou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zefeng Yang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Peipei He
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Jiachang Wu
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China,The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Luo J, Xia L, Zhang L, Zhao K, Li C. MiRNA-144-5p down-modulates CDCA3 to regulate proliferation and apoptosis of lung adenocarcinoma cells. Mutat Res 2022; 825:111798. [PMID: 36087462 DOI: 10.1016/j.mrfmmm.2022.111798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/24/2022] [Accepted: 08/28/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) belongs to non-small cell lung cancer. In addition to surgical resection, chemotherapy and radiotherapy cause great side effects and low 5-year survival rates. MiRNAs are closely related to cancer development. This study aimed to analyze the molecular mechanism of miRNA-144-5p targeting CDCA3 to inhibit LUAD proliferation. METHODS MiRNA and mRNA data were downloaded from TCGA-LUAD dataset for differential expression analysis. TargetScan and miRTarBase databases were adopted to predict the target genes of miRNA, and the signaling pathways involved were analyzed by gene set enrichment analysis. The functions of LUAD cells were analyzed by CCK-8, colony formation assay, stem cell spheroidization assay, and flow cytometry. The expression levels of CDCA3, p53, and cell cycle-associated proteins were evaluated by Western blot. RESULTS The expression of miRNA-144-5p was significantly down-regulated in LUAD, but overexpression of it repressed proliferation and spheroidization, and promoted apoptosis of LUAD cells. By bioinformatics prediction and dual-luciferase reporter assay, miRNA-144-5p was validated to target CDCA3, thereby regulating proliferation of LUAD cells. Besides, the results of cell experiments showed that miRNA-144-5p targeting CDCA3 affected cell proliferation and apoptosis in LUAD by regulating cell cycles, and miRNA-144-5p/CDCA3 mediated the p53 signaling pathway to affect the growth of LUAD cells. SIGNIFICANCE Through the study of the pathogenesis of miRNA-144-5p regulating LUAD, we can better understand the molecular mechanism underlying LUAD development.
Collapse
Affiliation(s)
- Jing Luo
- Thoracic Surgery, Zhejiang Hospital, Hangzhou, Zhejiang Province 310000, China
| | - Lilong Xia
- Thoracic Surgery, Zhejiang Hospital, Hangzhou, Zhejiang Province 310000, China
| | - Lei Zhang
- Thoracic Surgery, Zhejiang Hospital, Hangzhou, Zhejiang Province 310000, China
| | - Kaixiang Zhao
- Thoracic Surgery, Zhejiang Hospital, Hangzhou, Zhejiang Province 310000, China
| | - Chuanchuan Li
- Thoracic Surgery, Zhejiang Hospital, Hangzhou, Zhejiang Province 310000, China.
| |
Collapse
|
9
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
10
|
Zeng Z, Fei L, Yang J, Zuo J, Huang Z, Li H. MiR-27a-3p Targets GLP1R to Regulate Differentiation, Autophagy, and Release of Inflammatory Factors in Pre-Osteoblasts via the AMPK Signaling Pathway. Front Genet 2022; 12:783352. [PMID: 35069685 PMCID: PMC8766720 DOI: 10.3389/fgene.2021.783352] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/30/2021] [Indexed: 01/20/2023] Open
Abstract
Objective: Osteoporosis is caused by the dysregulation of bone homeostasis which is synergistically mediated by osteoclasts and osteoblasts. MiR-27a-3p is a key inhibitor of bone formation. Hence, unearthing the downstream target gene of miR-27a-3p is of great significance to understand the molecular mechanism of osteoporosis. Methods: Bioinformatics analysis was utilized to find the downstream target gene of miR-27a-3p, and dual-luciferase reporter assay was conducted to validate the interplay of miR-27a-3p and GLP1R. Besides, qRT-PCR, Western blot, and enzyme-linked immunosorbent assay (ELISA) were employed to verify the impact of miR-27a-3p on GLP1R expression and the differentiation, autophagy, and inflammatory response of MC3T3-E1 pre-osteoblasts. Results: Dual-luciferase assay validated that miR-27a-3p directly targeted GLP1R. Additionally, posttreatment of MC3T3-E1 cells with miR-27a-3p mimics resulted in a remarkable decrease in expression levels of GLP1R, cell differentiation marker gene, autophagy marker gene, and AMPK. These results indicated that miR-27a-3p targeted GLP1R to inhibit AMPK signal activation and pre-osteoblast differentiation and autophagy, while promoting the release of inflammatory factors. Conclusion: The miR-27a-3p/GLP1R regulatory axis in pre-osteoblasts contributes to understanding the molecular mechanism of osteoporosis.
Collapse
Affiliation(s)
- Zhi Zeng
- Department of Joint Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Liangyu Fei
- Department of Nephrology and Rheumatology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Juntao Yang
- Department of Hand and Foot Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jun Zuo
- Department of Hand and Foot Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zelin Huang
- Department of Hand and Foot Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Hao Li
- Department of Hand and Foot Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
11
|
Furesi G, de Jesus Domingues AM, Alexopoulou D, Dahl A, Hackl M, Schmidt JR, Kalkhof S, Kurth T, Taipaleenmäki H, Conrad S, Hofbauer C, Rauner M, Hofbauer LC. Exosomal miRNAs from Prostate Cancer Impair Osteoblast Function in Mice. Int J Mol Sci 2022; 23:1285. [PMID: 35163219 PMCID: PMC8836054 DOI: 10.3390/ijms23031285] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is the most frequent malignancy in older men with a high propensity for bone metastases. Characteristically, PCa causes osteosclerotic lesions as a result of disrupted bone remodeling. Extracellular vesicles (EVs) participate in PCa progression by conditioning the pre-metastatic niche. However, how EVs mediate the cross-talk between PCa cells and osteoprogenitors in the bone microenvironment remains poorly understood. We found that EVs derived from murine PCa cell line RM1-BM increased metabolic activity, vitality, and cell proliferation of osteoblast precursors by >60%, while significantly impairing mineral deposition (-37%). The latter was further confirmed in two complementary in vivo models of ossification. Accordingly, gene and protein set enrichments of osteoprogenitors exposed to EVs displayed significant downregulation of osteogenic markers and upregulation of proinflammatory factors. Additionally, transcriptomic profiling of PCa-EVs revealed the abundance of three microRNAs, miR-26a-5p, miR-27a-3p, and miR-30e-5p involved in the suppression of BMP-2-induced osteogenesis in vivo, suggesting the critical role of these EV-derived miRNAs in PCa-mediated suppression of osteoblast activity. Taken together, our results indicate the importance of EV cargo in cancer-bone cross-talk in vitro and in vivo and suggest that exosomal miRNAs may contribute to the onset of osteosclerotic bone lesions in PCa.
Collapse
Affiliation(s)
- Giulia Furesi
- Department of Medicine III & Center for Healthy Aging, Technical University of Dresden, 01307 Dresden, Germany; (G.F.); (S.C.); (M.R.)
| | | | - Dimitra Alexopoulou
- DRESDEN-Concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, 01307 Dresden, Germany; (D.A.); (A.D.)
| | - Andreas Dahl
- DRESDEN-Concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technical University of Dresden, 01307 Dresden, Germany; (D.A.); (A.D.)
| | | | - Johannes R. Schmidt
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology IZI, 04103 Leipzig, Germany; (J.R.S.); (S.K.)
| | - Stefan Kalkhof
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology IZI, 04103 Leipzig, Germany; (J.R.S.); (S.K.)
- Institute of Bioanalysis, University of Applied Sciences and Arts of Coburg, 96450 Coburg, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform, EM and Histology Facility, TU Dresden, 01307 Dresden, Germany;
| | - Hanna Taipaleenmäki
- Institute of Musculoskeletal Medicine (IMM), Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 82152 Planegg-Martinsried, Germany;
- Molecular Skeletal Biology Laboratory, Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefanie Conrad
- Department of Medicine III & Center for Healthy Aging, Technical University of Dresden, 01307 Dresden, Germany; (G.F.); (S.C.); (M.R.)
| | - Christine Hofbauer
- National Center for Tumor Diseases, Technical University of Dresden, 01307 Dresden, Germany;
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technical University of Dresden, 01307 Dresden, Germany; (G.F.); (S.C.); (M.R.)
| | - Lorenz C. Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technical University of Dresden, 01307 Dresden, Germany; (G.F.); (S.C.); (M.R.)
| |
Collapse
|
12
|
Liu Y, Fu D. lncRNA ZNF710-AS1 Acts as a ceRNA for miR-146a-5p and miR-146b-5p to Accelerate Osteogenic Differentiation of PDLSCs by Upregulating the BMP6/Smad1/5/9 Pathway. J HARD TISSUE BIOL 2022. [DOI: 10.2485/jhtb.31.231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ying Liu
- Dentistry Department, Jiufeng Street Health Service Center of East Lake Gaoxin District
| | - Dongjie Fu
- Department of Stomatology, Renmin Hospital of Wuhan University
| |
Collapse
|
13
|
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
|
14
|
Identification of Potential Osteoporosis miRNA Biomarkers Using Bioinformatics Approaches. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:3562942. [PMID: 34777562 PMCID: PMC8579105 DOI: 10.1155/2021/3562942] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/27/2021] [Indexed: 11/17/2022]
Abstract
Osteoporosis is a degenerative osteoarthropathy commonly found in old people and postmenopausal women. Many studies showed that microRNAs (miRNAs) can regulate the expression of osteoporosis-related genes and are abnormally expressed in patients with osteoporosis. miRNAs therefore have the potential to serve as biomarkers of osteoporosis. In this study, the limma package was used for the differential expression analysis of mRNA expression profiles and 357 significantly differentially expressed genes (DEGs) were obtained. Metascape was used for functional enrichment analysis of DEGs. The result revealed that DEGs were mainly enriched in signaling pathways like MAPK6/MAPK4. Based on the STRING database, the protein-protein interaction (PPI) network of DEGs was constructed. MCODE was used to analyze the functional subsets, and a key functional subset composed of 9 genes was screened out. In addition, the miRNA-mRNA regulatory interaction network (RegIN) was analyzed by the CyTargetLinker plugin, which generated 55 miRNA-mRNA regulatory interactions. Through literature searching, the osteoporosis-related gene FOXO1 in the key functional subset was determined to be the main object of the study. In qRT-PCR assay, the expression of the predicted miRNAs was tested in peripheral blood mononuclear cells of mice with osteoporosis, in which 13 miRNAs were remarkably highly expressed. All in all, this study, based on bioinformatics analysis and testing assay of miRNA expression, determined the potential biomarkers of osteoporosis.
Collapse
|
15
|
Han X, Fan Z. MicroRNAs Regulation in Osteogenic Differentiation of Mesenchymal Stem Cells. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.747068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a kind of pluripotent stem cell with the potential of self-renewal and multidirectional differentiation. They can be obtained from a variety of tissues and can differentiate into a variety of cell types under different induction conditions, including osteoblasts. Because of this osteogenic property, MSCs have attracted much attention in the treatment of bone metabolism-related diseases. MicroRNAs (miRNAs), as an epigenetic factor, are thought to play an important regulatory role in the process of osteogenic differentiation of MSCs. In recent years, increasingly evidence shows that miRNAs imbalance is involved in the regulation of osteoporosis and fracture. In this review, miRNAs involved in osteogenic differentiation and their mechanisms for regulating the expression of target genes are reviewed. In addition, we also discuss the potential clinical applications and possible directions of this field in the future.
Collapse
|
16
|
Ghafouri-Fard S, Abak A, Tavakkoli Avval S, Rahmani S, Shoorei H, Taheri M, Samadian M. Contribution of miRNAs and lncRNAs in osteogenesis and related disorders. Biomed Pharmacother 2021; 142:111942. [PMID: 34311172 DOI: 10.1016/j.biopha.2021.111942] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/07/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Non-coding RNAs have been found to regulate several developmental processes among them is osteogenesis. Although these transcripts have several distinct classes, two classes i.e. microRNAs and long non-coding RNAs have attained more attention. These transcripts regulate intramembranous as well as endochondral ossification processes. The effects of microRNAs on osteogenesis are mostly mediated through modulation of Wnt/β-catenin and TGFβ/BMP pathways. Long non-coding RNAs can directly affect expression of these pathways or osteogenic transcription factors. Moreover, they can serve as a molecular sponge for miRNAs. MALAT1/miR-30, MALAt1/miR-214, LEF1-AS1/miR-24-3p, MCF2L-AS1/miR-33a, MSC-AS1/miR-140-5p and KCNQ1OT1/miR-214 are examples of such kind of interaction between lncRNAs and miRNAs in the context of osteogenesis. In the current paper, we explain these two classes of non-coding RNAs in the osteogenesis and related disorders.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shayan Rahmani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Samadian
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
17
|
Zhang YL, Liu L, Peymanfar Y, Anderson P, Xian CJ. Roles of MicroRNAs in Osteogenesis or Adipogenesis Differentiation of Bone Marrow Stromal Progenitor Cells. Int J Mol Sci 2021; 22:ijms22137210. [PMID: 34281266 PMCID: PMC8269269 DOI: 10.3390/ijms22137210] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022] Open
Abstract
Bone marrow stromal cells (BMSCs) are multipotent cells which can differentiate into chondrocytes, osteoblasts, and fat cells. Under pathological stress, reduced bone formation in favour of fat formation in the bone marrow has been observed through a switch in the differentiation of BMSCs. The bone/fat switch causes bone growth defects and disordered bone metabolism in bone marrow, for which the mechanisms remain unclear, and treatments are lacking. Studies suggest that small non-coding RNAs (microRNAs) could participate in regulating BMSC differentiation by disrupting the post-transcription of target genes, leading to bone/fat formation changes. This review presents an emerging concept of microRNA regulation in the bone/fat formation switch in bone marrow, the evidence for which is assembled mainly from in vivo and in vitro human or animal models. Characterization of changes to microRNAs reveals novel networks that mediate signalling and factors in regulating bone/fat switch and homeostasis. Recent advances in our understanding of microRNAs in their control in BMSC differentiation have provided valuable insights into underlying mechanisms and may have significant potential in development of new therapeutics.
Collapse
|
18
|
Wu YZ, Huang HT, Cheng TL, Lu YM, Lin SY, Ho CJ, Lee TC, Hsu CH, Huang PJ, Huang HH, Li JY, Su YD, Chen SC, Kang L, Chen CH. Application of microRNA in Human Osteoporosis and Fragility Fracture: A Systemic Review of Literatures. Int J Mol Sci 2021; 22:ijms22105232. [PMID: 34063380 PMCID: PMC8156577 DOI: 10.3390/ijms22105232] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) could serve as ideal entry points to the deregulated pathways in osteoporosis due to their relatively simple upstream and downstream relationships with other molecules in the signaling cascades. Our study aimed to give a comprehensive review of the already identified miRNAs in osteoporosis from human blood samples and provide useful information for their clinical application. A systematic literature search for relevant studies was conducted in the Pubmed database from inception to December 2020. We set two essential inclusion criteria: human blood sampling and design of controlled studies. We sorted the results of analysis on human blood samples according to the study settings and compiled the most promising miRNAs with analyzed diagnostic values. Furthermore, in vitro and in vivo evidence for the mechanisms of the identified miRNAs was also illustrated. Based on both diagnostic value and evidence of mechanism from in vitro and in vivo experiments, miR-23b-3p, miR-140-3p, miR-300, miR-155-5p, miR-208a-3p, and miR-637 were preferred candidates in diagnostic panels and as therapeutic agents. Further studies are needed to build sound foundations for the clinical usage of miRNAs in osteoporosis.
Collapse
Affiliation(s)
- Yen-Zung Wu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Hsuan-Ti Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Tsung-Lin Cheng
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Yen-Mou Lu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Cheng-Jung Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Tien-Ching Lee
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Chia-Hao Hsu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Peng-Ju Huang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Han Hsiang Huang
- Department of Veterinary Medicine, National Chiayi University, Chiayi 60004, Taiwan;
| | - Jhong-You Li
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Yu-De Su
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan
| | - Shih-Chieh Chen
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Department of Medical Records, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: (L.K.); (C.-H.C.); Tel.: +886-7-3209-209 (C.-H.C.)
| | - Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan; (Y.-Z.W.); (H.-T.H.); (T.-L.C.); (Y.-M.L.); (S.-Y.L.); (C.-J.H.); (T.-C.L.); (C.-H.H.); (P.-J.H.); (J.-Y.L.); (Y.-D.S.)
- Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Regeneration Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Musculoskeletal Regeneration Research Center, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, College of Medicine, Kaohsiung Medical University, Kaohsiung 80701, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Healthcare Administration and Medical Informatics, Kaohsiung Medical University, Kaohsiung 80701, Taiwan;
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 80420, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912301, Taiwan
- Correspondence: (L.K.); (C.-H.C.); Tel.: +886-7-3209-209 (C.-H.C.)
| |
Collapse
|
19
|
Grillari J, Mäkitie RE, Kocijan R, Haschka J, Vázquez DC, Semmelrock E, Hackl M. Circulating miRNAs in bone health and disease. Bone 2021; 145:115787. [PMID: 33301964 DOI: 10.1016/j.bone.2020.115787] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022]
Abstract
microRNAs have evolved as important regulators of multiple biological pathways essential for bone homeostasis, and microRNA research has furthered our understanding of the mechanisms underlying bone health and disease. This knowledge, together with the finding that active or passive release of microRNAs from cells into the extracellular space enables minimal-invasive detection in biofluids (circulating miRNAs), motivated researchers to explore microRNAs as biomarkers in several pathologic conditions, including bone diseases. Thus, exploratory studies in cohorts representing different types of bone diseases have been performed. In this review, we first summarize important molecular basics of microRNA function and release and provide recommendations for best (pre-)analytical practices and documentation standards for circulating microRNA research required for generating high quality data and ensuring reproducibility of results. Secondly, we review how the genesis of bone-derived circulating microRNAs via release from osteoblasts and osteoclasts could contribute to the communication between these cells. Lastly, we summarize evidence from clinical research studies that have investigated the clinical utility of microRNAs as biomarkers in musculoskeletal disorders. While previous reviews have mainly focused on diagnosis of primary osteoporosis, we have also included studies exploring the utility of circulating microRNAs in monitoring anti-osteoporotic treatment and for diagnosis of other types of bone diseases, such as diabetic osteopathy, bone degradation in inflammatory diseases, and monogenetic bone diseases.
Collapse
Affiliation(s)
- Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria; Institute for Molecular Biotechnology, BOKU - University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Riikka E Mäkitie
- Folkhälsan Institute of Genetics and University of Helsinki, Helsinki, Finland; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Molecular Endocrinology Laboratory, Department of Medicine, Hammersmith Campus, Imperial College London, London, United Kingdom
| | - Roland Kocijan
- Hanusch Hospital of the WGKK and AUVA Trauma Center, 1st Medical Department at Hanusch Hospital, Ludwig Boltzmann Institute of Osteology, Vienna, Austria; Sigmund Freud University Vienna, School of Medicine, Metabolic Bone Diseases Unit, Austria
| | - Judith Haschka
- Hanusch Hospital of the WGKK and AUVA Trauma Center, 1st Medical Department at Hanusch Hospital, Ludwig Boltzmann Institute of Osteology, Vienna, Austria; Karl Landsteiner Institute for Rheumatology and Gastroenterology, Vienna, Austria
| | | | | | - Matthias Hackl
- Austrian Cluster for Tissue Regeneration, Austria; TAmiRNA GmbH, Vienna, Austria.
| |
Collapse
|
20
|
Kinget L, Roussel E, Lambrechts D, Boeckx B, Vanginderhuysen L, Albersen M, Rodríguez-Antona C, Graña-Castro O, Inglada-Pérez L, Verbiest A, Zucman-Rossi J, Couchy G, Caruso S, Laenen A, Baldewijns M, Beuselinck B. MicroRNAs Possibly Involved in the Development of Bone Metastasis in Clear-Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13071554. [PMID: 33800656 PMCID: PMC8036650 DOI: 10.3390/cancers13071554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Bone metastases cause substantial morbidity and implicate worse clinical outcomes for clear-cell renal cell carcinoma patients. MicroRNAs are small RNA molecules that modulate gene translation and are involved in the development of cancer and metastasis. We identified six microRNAs that are potentially specifically involved in metastasis to bone, of which two seem protective and four implicate a higher risk. This aids further understanding of the process of metastasizing to bone. Furthermore, these microRNA hold potential for biomarkers or therapeutic targets. Abstract Bone metastasis in clear-cell renal cell carcinoma (ccRCC) leads to substantial morbidity through skeletal related adverse events and implicates worse clinical outcomes. MicroRNAs (miRNA) are small non-protein coding RNA molecules with important regulatory functions in cancer development and metastasis. In this retrospective analysis we present dysregulated miRNA in ccRCC, which are associated with bone metastasis. In particular, miR-23a-3p, miR-27a-3p, miR-20a-5p, and miR-335-3p specifically correlated with the earlier appearance of bone metastasis, compared to metastasis in other organs. In contrast, miR-30b-3p and miR-139-3p were correlated with less occurrence of bone metastasis. These miRNAs are potential biomarkers and attractive targets for miRNA inhibitors or mimics, which could lead to novel therapeutic possibilities for bone targeted treatment in metastatic ccRCC.
Collapse
Affiliation(s)
- Lisa Kinget
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (E.R.); (M.A.)
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (D.L.); (B.B.)
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium; (D.L.); (B.B.)
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Loïc Vanginderhuysen
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium; (E.R.); (M.A.)
| | | | - Osvaldo Graña-Castro
- Centro Nacional de Investigaciones Oncológicas (CNIO), 28040 Madrid, Spain; (C.R.-A.); (O.G.-C.)
| | - Lucía Inglada-Pérez
- Department of Statistics and Operational Research, Faculty of Medicine, Complutense University, 28040 Madrid, Spain;
| | - Annelies Verbiest
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | - Gabrielle Couchy
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, INSERM, Functional Genomics of Solid Tumors Laboratory, Équipe Labellisée Ligue Nationale contre le Cancer, Labex OncoImmunology, F-75006 Paris, France; (J.Z.-R.); (G.C.); (S.C.)
| | | | | | - Benoit Beuselinck
- Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, 3000 Leuven, Belgium; (L.K.); (L.V.); (A.V.)
- Correspondence: ; Tel.: +32-16-346900
| |
Collapse
|