1
|
Huang Y, Wang Z. Therapeutic potential of SOX family transcription factors in osteoarthritis. Ann Med 2025; 57:2457520. [PMID: 39887675 PMCID: PMC11789227 DOI: 10.1080/07853890.2025.2457520] [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: 06/22/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND As the worldwide population ages, osteoarthritis has significantly increased. This musculoskeletal condition has become a pressing global health issue and thus, prevention and treatment of osteoarthritis have become the primary focus of domestic and international research. Scholarly investigations of the molecular mechanisms that are related to the occurrence and development of osteoarthritis have shed light on the pathological causes of this condition to a certain extent, providing a foundation for its prevention and treatment. However, further research is necessary to fully understand the critical role of the transcription factor SOX9 in chondrocyte differentiation and the development of osteoarthritis. As a result, there has been widespread interest in SOX transcription factors. While SOX9 has been utilized as a biomarker to indicate the occurrence and prognosis of osteoarthritis, investigations into other members of the SOX family and the development of targeted treatments around SOX9 are still required. PURPOSE This article considers the impact of the SOX protein on the development and inhibition of osteoarthritis and highlights the need for therapeutic approaches targeting SOX9, as supported by existing research. RESULTS SOX9 can contribute to the process of osteoarthritis through acetylation and ubiquitination modifications. The regulation of the WNT signalling pathway, Nrf2/ARE signalling pathway, NF-κB signalling pathway and SOX9 is implicated in the emergence of osteoarthritis. Non-coding RNA may play a role in the onset and progression of osteoarthritis by modulating various SOX family members, including SOX2, SOX4, SOX5, SOX6, SOX8, SOX9 and SOX11. CONCLUSION SOX9 has the capability of mitigating the onset and progression of osteoarthritis through means such as medication therapy, stem cell therapy, recombinant adeno-associated virus (rAAV) vector therapy, physical therapy and other approaches.
Collapse
Affiliation(s)
- Yue Huang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
2
|
Lin N, Song Z, Ma B, Wang P. LncRNA SNHG7 inhibits apoptosis and proliferation of osteoarthritis cells induced by IL-β through sponging miR-146b. Connect Tissue Res 2025:1-14. [PMID: 40207563 DOI: 10.1080/03008207.2025.2487470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/24/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE We probed the roles of SNHG7, miR-146b, PCBP1, and IL-β in the development of osteoarthritis (OA). MATERIALS AND METHODS OA models were established using anterior cruciate ligaments, and chondrocytes were obtained from mouse cartilage tissue. Cells were treated with 10 ng/ml Il-1β. RT-qPCR was used to detect the expression of SNHG7, miR-146b, PCBP1, and IL-β in tissues and cells. Safranin-O/Fast Green staining was performed to analyze the cartilage damage in each group of mice. RESULTS SNHG7 and PCBP1 expressions were down-regulated, and miR-146b expression was up-regulated in OA tissue and IL-1β-treated chondrocytes compared to normal cartilage tissue and chondrocytes. Forced SNHG7 expression improved cartilage structure, enhanced proliferative viability of chondrocytes, and inhibited apoptosis and IL-1β release in IL-1β-treated chondrocytes in OA mice. In contrast, miR-146b upregulation decreased proliferative viability and promoted apoptosis and IL-1β release in chondrocytes. Rescue assays showed that miR-146b attenuated the protective effects of SNHG7 on apoptosis and inflammation in IL-1β-treated chondrocytes, and activation of PCBP1 expression significantly inhibited the cytotoxic effects of miR-146b. Mechanistically, SNHG7 acted as a competitive endogenous RNA by targeting miR-146b to promote the expression of PCBP1. CONCLUSIONS This study confirms that SNHG7 inhibits IL-1β-mediated inflammatory responses in chondrocytes via the miR-146b/PCBP1 axis, thereby suppressing IL-1β-induced OA.
Collapse
Affiliation(s)
- Naikai Lin
- Department of Orthopedics, Hangzhou Yuhang District First People's Hospital, Zhejiang, China
| | - Zehui Song
- Department of Orthopedics, Hangzhou Yuhang District First People's Hospital, Zhejiang, China
| | - Bitao Ma
- Department of Traditional Chinese Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peng Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Villagrán-Andrade KM, Núñez-Carro C, Blanco FJ, de Andrés MC. Nutritional Epigenomics: Bioactive Dietary Compounds in the Epigenetic Regulation of Osteoarthritis. Pharmaceuticals (Basel) 2024; 17:1148. [PMID: 39338311 PMCID: PMC11434976 DOI: 10.3390/ph17091148] [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: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Nutritional epigenomics is exceptionally important because it describes the complex interactions among food compounds and epigenome modifications. Phytonutrients or bioactive compounds, which are secondary metabolites of plants, can protect against osteoarthritis by suppressing the expression of inflammatory and catabolic mediators, modulating epigenetic changes in DNA methylation, and the histone or chromatin remodelling of key inflammatory genes and noncoding RNAs. The combination of natural epigenetic modulators is crucial because of their additive and synergistic effects, safety and therapeutic efficacy, and lower adverse effects than conventional pharmacology in the treatment of osteoarthritis. In this review, we have summarized the chondroprotective properties of bioactive compounds used for the management, treatment, or prevention of osteoarthritis in both human and animal studies. However, further research is needed into bioactive compounds used as epigenetic modulators in osteoarthritis, in order to determine their potential value for future clinical applications in osteoarthritic patients as well as their relation with the genomic and nutritional environment, in order to personalize food and nutrition together with disease prevention.
Collapse
Affiliation(s)
- Karla Mariuxi Villagrán-Andrade
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Carmen Núñez-Carro
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Francisco J Blanco
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación en Reumatología y Salud, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - María C de Andrés
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| |
Collapse
|
4
|
Shakeri M, Aminian A, Mokhtari K, Bahaeddini M, Tabrizian P, Farahani N, Nabavi N, Hashemi M. Unraveling the molecular landscape of osteoarthritis: A comprehensive review focused on the role of non-coding RNAs. Pathol Res Pract 2024; 260:155446. [PMID: 39004001 DOI: 10.1016/j.prp.2024.155446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024]
Abstract
Osteoarthritis (OA) poses a significant global health challenge, with its prevalence anticipated to increase in the coming years. This review delves into the emerging molecular biomarkers in OA pathology, focusing on the roles of various molecules such as metabolites, noncoding RNAs (ncRNAs), including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). Advances in omics technologies have transformed biomarker identification, enabling comprehensive analyses of the complex pathways involved in OA pathogenesis. Notably, ncRNAs, especially miRNAs and lncRNAs, exhibit dysregulated expression patterns in OA, presenting promising opportunities for diagnosis and therapy. Additionally, the intricate interplay between epigenetic modifications and OA progression highlights the regulatory role of epigenetics in gene expression dynamics. Genome-wide association studies have pinpointed key genes undergoing epigenetic changes, providing insights into the inflammatory processes and chondrocyte hypertrophy typical of OA. Understanding the molecular landscape of OA, including biomarkers and epigenetic mechanisms, holds significant potential for developing innovative diagnostic tools and therapeutic strategies for OA management.
Collapse
Affiliation(s)
- Mohammadreza Shakeri
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Aminian
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mohammadreza Bahaeddini
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Pouria Tabrizian
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
5
|
Giammona A, Di Franco S, Lo Dico A, Stassi G. The miRNA Contribution in Adipocyte Maturation. Noncoding RNA 2024; 10:35. [PMID: 38921832 PMCID: PMC11206860 DOI: 10.3390/ncrna10030035] [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: 04/15/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Mesenchymal stem cells, due to their multipotent ability, are considered one of the best candidates to be used in regenerative medicine. To date, the most used source is represented by the bone marrow, despite the limited number of cells and the painful/invasive procedure for collection. Therefore, the scientific community has investigated many alternative sources for the collection of mesenchymal stem cells, with the adipose tissue representing the best option, given the abundance of mesenchymal stem cells and the easy access. Although adipose mesenchymal stem cells have recently been investigated for their multipotency, the molecular mechanisms underlying their adipogenic potential are still unclear. In this scenario, this communication is aimed at defining the role of miRNAs in adipogenic potential of adipose-derived mesenchymal stem cells via real-time PCR. Even if preliminary, our data show that cell culture conditions affect the expression of specific miRNA involved in the adipogenic potential of mesenchymal stem cells. The in vitro/in vivo validation of these results could pave the way for novel therapeutic strategies in the field of regenerative medicine. In conclusion, our research highlights how specific cell culture conditions can modulate the adipogenic potential of adipose mesenchymal stem cells through the regulation of specific miRNAs.
Collapse
Affiliation(s)
- Alessandro Giammona
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), 20054 Segrate, Italy;
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Laboratory of Cellular and Molecular Pathophysiology, Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy;
| | - Simone Di Franco
- Laboratory of Cellular and Molecular Pathophysiology, Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy;
| | - Alessia Lo Dico
- Institute of Molecular Bioimaging and Physiology (IBFM), National Research Council (CNR), 20054 Segrate, Italy;
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Giorgio Stassi
- Laboratory of Cellular and Molecular Pathophysiology, Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, 90127 Palermo, Italy;
| |
Collapse
|
6
|
Dalle Carbonare L, Minoia A, Braggio M, Bertacco J, Piritore FC, Zouari S, Vareschi A, Elia R, Vedovi E, Scumà C, Carlucci M, Bhandary L, Mottes M, Romanelli MG, Valenti MT. Modulation of miR-146b Expression during Aging and the Impact of Physical Activity on Its Expression and Chondrogenic Progenitors. Int J Mol Sci 2023; 24:13163. [PMID: 37685971 PMCID: PMC10488278 DOI: 10.3390/ijms241713163] [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: 08/03/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The finding of molecules associated with aging is important for the prevention of chronic degenerative diseases and for longevity strategies. MicroRNAs (miRNAs) are post-transcriptional regulators involved in many biological processes and miR-146b-5p has been shown to be involved in different degenerative diseases. However, miR-146b-5p modulation has not been evaluated in mesenchymal stem cells (MSCs) commitment or during aging. Therefore, the modulation of miR-146b-5p in the commitment and differentiation of mesenchymal cells as well as during maturation and aging in zebrafish model were analyzed. In addition, circulating miR-146b-5p was evaluated in human subjects at different age ranges. Thus, the role of physical activity in the modulation of miR-146b-5p was also investigated. To achieve these aims, RT (real-time)-PCR, Western blot, cell transfections, and three-dimensional (3D) culture techniques were applied. Our findings show that miR-146b-5p expression drives MSCs to adipogenic differentiation and increases during zebrafish maturation and aging. In addition, miR-146b-5p expression is higher in females compared to males and it is associated with the aging in humans. Interestingly, we also observed that the physical activity of walking downregulates circulating miR-146b-5p levels in human females and increases the number of chondroprogenitors. In conclusion, miR-146b-5p can be considered an age-related marker and can represent a useful marker for identifying strategies, such as physical activity, aimed at counteracting the degenerative processes of aging.
Collapse
Affiliation(s)
- Luca Dalle Carbonare
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Arianna Minoia
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Michele Braggio
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Jessica Bertacco
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Francesca Cristiana Piritore
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Sharazed Zouari
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Anna Vareschi
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Rossella Elia
- Department of Engineering for Innovative Medicine, University of Verona, 37100 Verona, Italy; (L.D.C.); (A.M.); (M.B.); (S.Z.); (A.V.); (R.E.)
| | - Ermes Vedovi
- Recovery and Functional Rehabilitation, Integrated University Hospital of Verona, 37100 Verona, Italy; (E.V.); (C.S.)
| | - Cristina Scumà
- Recovery and Functional Rehabilitation, Integrated University Hospital of Verona, 37100 Verona, Italy; (E.V.); (C.S.)
| | - Matilde Carlucci
- Health Directorate, Integrated University Hospital of Verona, 37100 Verona, Italy;
| | | | - Monica Mottes
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Maria Grazia Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| | - Maria Teresa Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100 Verona, Italy; (J.B.); (F.C.P.); (M.M.); (M.G.R.)
| |
Collapse
|
7
|
Muñoz-García R, Sánchez-Hidalgo M, Alcarranza M, Vazquéz-Román MV, de Sotomayor MA, González-Rodríguez ML, de Andrés MC, Alarcón-de-la-Lastra C. Effects of Dietary Oleacein Treatment on Endothelial Dysfunction and Lupus Nephritis in Balb/C Pristane-Induced Mice. Antioxidants (Basel) 2023; 12:1303. [PMID: 37372034 DOI: 10.3390/antiox12061303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic immune-inflammatory disease characterized by multiorgan affectation and lowered self-tolerance. Additionally, epigenetic changes have been described as playing a pivotal role in SLE. This work aims to assess the effects of oleacein (OLA), one of the main extra virgin olive oil secoiridoids, when used to supplement the diet of a murine pristane-induced SLE model. In the study, 12-week-old female BALB/c mice were injected with pristane and fed with an OLA-enriched diet (0.01 % (w/w)) for 24 weeks. The presence of immune complexes was evaluated by immunohistochemistry and immunofluorescence. Endothelial dysfunction was studied in thoracic aortas. Signaling pathways and oxidative-inflammatory-related mediators were evaluated by Western blotting. Moreover, we studied epigenetic changes such as DNA methyltransferase (DNMT-1) and micro(mi)RNAs expression in renal tissue. Nutritional treatment with OLA reduced the deposition of immune complexes, ameliorating kidney damage. These protective effects could be related to the modulation of mitogen-activated protein kinases, the Janus kinase/signal transducer and transcription activator of transcription, nuclear factor kappa, nuclear-factor-erythroid-2-related factor 2, inflammasome signaling pathways, and the regulation of miRNAs (miRNA-126, miRNA-146a, miRNA-24-3p, and miRNA-123) and DNMT-1 expression. Moreover, the OLA-enriched diet normalized endothelial nitric oxide synthase and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-1 overexpression. These preliminary results suggest that an OLA-supplemented diet could constitute a new alternative nutraceutical therapy in the management of SLE, supporting this compound as a novel epigenetic modulator of the immunoinflammatory response.
Collapse
Affiliation(s)
- Rocío Muñoz-García
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
| | - Marina Sánchez-Hidalgo
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
| | - Manuel Alcarranza
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
| | - María Victoria Vazquéz-Román
- Department of Normal and Pathological Cytology and Histology, Faculty of Medicine, Universidad de Sevilla, 41012 Seville, Spain
| | | | | | - María C de Andrés
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Catalina Alarcón-de-la-Lastra
- Department of Pharmacology, Faculty of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain
| |
Collapse
|
8
|
Yassin AM, AbuBakr HO, Abdelgalil AI, Farid OA, El-Behairy AM, Gouda EM. Circulating miR-146b and miR-27b are efficient biomarkers for early diagnosis of Equidae osteoarthritis. Sci Rep 2023; 13:7966. [PMID: 37198318 PMCID: PMC10192321 DOI: 10.1038/s41598-023-35207-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/14/2023] [Indexed: 05/19/2023] Open
Abstract
One of the most orthopedic problems seen in the equine is osteoarthritis (OA). The present study tracks some biochemical, epigenetic, and transcriptomic factors along different stages of monoiodoacetate (MIA) induced OA in donkeys in serum and synovial fluid. The aim of the study was the detection of sensitive noninvasive early biomarkers. OA was induced by a single intra-articular injection of 25 mg of MIA into the left radiocarpal joint of nine donkeys. Serum and synovial samples were taken at zero-day and different intervals for assessment of total GAGs and CS levels as well as miR-146b, miR-27b, TRAF-6, and COL10A1 gene expression. The results showed that the total GAGs and CS levels increased in different stages of OA. The level of expression of both miR-146b and miR-27b were upregulated as OA progressed and then downregulated at late stages. TRAF-6 gene was upregulated at the late stage while synovial fluid COL10A1 was over-expressed at the early stage of OA and then decreased at the late stages (P < 0.05). In conclusion, both miR-146b and miR-27b together with COL10A1 could be used as promising noninvasive biomarkers for the very early diagnosis of OA.
Collapse
Affiliation(s)
- Aya M Yassin
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt.
| | - Huda O AbuBakr
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Ahmed I Abdelgalil
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Omar A Farid
- Department of Physiology, National Organization for Drug Control and Research, Giza, Egypt
| | - Adel M El-Behairy
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Eman M Gouda
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| |
Collapse
|
9
|
Núñez-Carro C, Blanco-Blanco M, Villagrán-Andrade KM, Blanco FJ, de Andrés MC. Epigenetics as a Therapeutic Target in Osteoarthritis. Pharmaceuticals (Basel) 2023; 16:156. [PMID: 37259307 PMCID: PMC9964205 DOI: 10.3390/ph16020156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 08/15/2023] Open
Abstract
Osteoarthritis (OA) is a heterogenous, complex disease affecting the integrity of diarthrodial joints that, despite its high prevalence worldwide, lacks effective treatment. In recent years it has been discovered that epigenetics may play an important role in OA. Our objective is to review the current knowledge of the three classical epigenetic mechanisms-DNA methylation, histone post-translational modifications (PTMs), and non-coding RNA (ncRNA) modifications, including microRNAs (miRNAs), circular RNAs (circRNAs), and long non-coding RNAs (lncRNAs)-in relation to the pathogenesis of OA and focusing on articular cartilage. The search for updated literature was carried out in the PubMed database. Evidence shows that dysregulation of numerous essential cartilage molecules is caused by aberrant epigenetic regulatory mechanisms, and it contributes to the development and progression of OA. This offers the opportunity to consider new candidates as therapeutic targets with the potential to attenuate OA or to be used as novel biomarkers of the disease.
Collapse
Affiliation(s)
- Carmen Núñez-Carro
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Margarita Blanco-Blanco
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Karla Mariuxi Villagrán-Andrade
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| | - Francisco J. Blanco
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
- Grupo de Investigación en Reumatología y Salud, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Campus de Oza, Universidade da Coruña (UDC), 15008 A Coruña, Spain
| | - María C. de Andrés
- Unidad de Epigenética, Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario, de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain
| |
Collapse
|
10
|
Regenerative mesenchymal stem c
ell‐derived
extracellular vesicles: A potential alternative to c
ell‐based
therapy in viral infection and disease damage control. WIREs Mech Dis 2022; 14:e1574. [DOI: 10.1002/wsbm.1574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/24/2022] [Indexed: 11/07/2022]
|
11
|
Feng L, Yang Z, Li Y, Pan Q, Zhang X, Wu X, Lo JHT, Wang H, Bai S, Lu X, Wang M, Lin S, Pan X, Li G. MicroRNA-378 contributes to osteoarthritis by regulating chondrocyte autophagy and bone marrow mesenchymal stem cell chondrogenesis. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:328-341. [PMID: 35474736 PMCID: PMC9010521 DOI: 10.1016/j.omtn.2022.03.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/17/2022] [Indexed: 12/21/2022]
Abstract
Osteoarthritis (OA) is the most common joint disease; thus, understanding the pathological mechanisms of OA initiation and progression is critical for OA treatment. MicroRNAs (miRNAs) have been shown to be involved in the progression of osteoarthritis, one candidate is microRNA-378 (miR-378), which is highly expressed in the synovium of OA patients during late-stage disease, but its function and the underlying mechanisms of how it contributes to disease progression remain poorly understood. In this study, miR-378 transgenic (TG) mice were used to study the role of miR-378 in OA development. miR-378 TG mice developed spontaneous OA and also exaggerated surgery-induced disease progression. Upon in vitro OA induction, miR-378 expression was upregulated and correlated with elevated inflammation and chondrocyte hypertrophy. Chondrocytes isolated from articular cartilage from miR-378 TG mice showed impaired chondrogenic differentiation. The bone marrow mesenchymal stem cells (BMSCs) collected from miR-378 TG mice also showed repressed chondrogenesis compared with the control group. The autophagy-related protein Atg2a, as well as chondrogenesis regulator Sox6, were identified as downstream targets of miR-378. Ectopic expression of Atg2a and Sox6 rescued miR-378-repressed chondrocyte autophagy and BMSC chondrogenesis, respectively. Anti-miR-378 lentivirus intra-articular injection in an established OA mouse model was shown to ameliorate OA progression, promote articular regeneration, and repress hypertrophy. Atg2a and Sox6 were again confirmed to be the target of miR-378 in vivo. In conclusion, miR-378 amplified OA development via repressing chondrocyte autophagy and by inhibiting BMSCs chondrogenesis, thus indicating miR-378 may be a potential therapeutic target for OA treatments.
Collapse
Affiliation(s)
- Lu Feng
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Zhengmeng Yang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Yucong Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Qi Pan
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
- Department of Pediatric Orthopaedics, South China Hospital, Health Science Center, Shenzhen University, Shenzhen 518116, PR China
| | - Xiaoting Zhang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Xiaomin Wu
- Department of Orthopaedics and Traumatology, People’s Hospital of Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| | - Jessica Hiu Tung Lo
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Haixing Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Shanshan Bai
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Xuan Lu
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Ming Wang
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | - Xiaohua Pan
- Department of Orthopaedics and Traumatology, People’s Hospital of Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| |
Collapse
|
12
|
Liu JN, Lu S, Fu CM. MiR-146a expression profiles in osteoarthritis in different tissue sources: a meta-analysis of observational studies. J Orthop Surg Res 2022; 17:148. [PMID: 35248106 PMCID: PMC8898505 DOI: 10.1186/s13018-022-02989-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023] Open
Abstract
Background MiR-146a has been widely studied in the pathogenesis of osteoarthritis (OA); however, the results are still controversial. Objective This meta-analysis analyzes the expression profile of miR-146a in various tissues of OA patients. Methods Public databases were searched for appropriate studies published up to September 1, 2021. A case–control study comparing the OA population and a non-OA healthy population was included. Results 26 articles were included in analysis. The results showed that the expression level of miR-146a in peripheral blood mononuclear cells (PBMCs) was significantly higher in OA patients than in controls (SMD: 1.23; 95% CI 0.08–2.37; p = 0.035) but not in plasma (SMD: 1.09; 95% CI − 0.06, 2.24; p = 0.064). The expression level of miR-146a in cartilage was also significantly higher in OA patients than in controls (SMD: 6.39; 95% CI 0.36, 12.4; p = 0.038) but not in chondrocytes (SMD: − 0.71; 95% CI − 4.15, 2.73; p = 0.687). The miR-146a level was significantly lower in synoviocytes in the OA population than in control patients (SMD: − 0.97; 95% CI − 1.68, − 0.26; p = 0.008). In synovial tissue, synovial fluid, and regulatory T cells, there was no significant difference. Conclusion The expression level of miR-146a in cartilage tissue and PBMCs was significantly higher in OA patients than in non-OA healthy controls. Due to the limitations of this study, more research is needed to confirm these results in the future. Trial registration: retrospectively registered. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-022-02989-7.
Collapse
|
13
|
Lie MU, Pedersen LM, Heuch I, Winsvold B, Gjerstad J, Hasvik E, Nygaard ØP, Grotle M, Matre D, Zwart JA, Nilsen KB. Low Back Pain With Persistent Radiculopathy; the Clinical Role of Genetic Variants in the Genes SOX5, CCDC26/GSDMC and DCC. Front Genet 2022; 12:757632. [PMID: 35140737 PMCID: PMC8819060 DOI: 10.3389/fgene.2021.757632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/29/2021] [Indexed: 12/28/2022] Open
Abstract
In a recently published genome-wide association study (GWAS) chronic back pain was associated with three loci; SOX5, CCDC26/GSDMC and DCC. This GWAS was based on a heterogeneous sample of back pain disorders, and it is unknown whether these loci are of clinical relevance for low back pain (LBP) with persistent radiculopathy. Thus, we examine if LBP with radiculopathy 12 months after an acute episode of LBP with radiculopathy is associated with the selected single nucleotide polymorphisms (SNPs); SOX5 rs34616559, CCDC26/GSDMC rs7833174 and DCC rs4384683. In this prospective cohort study, subjects admitted to a secondary health care institution due to an acute episode of LBP with radiculopathy, reported back pain, leg pain, and Oswestry Disability Index (ODI), were genotyped and followed up at 12 months (n = 338). Kruskal-Wallis H test showed no association between the SNPs and back pain, leg pain or ODI. In conclusion, LBP with radiculopathy 12 months after an acute episode of LBP with radiculopathy, is not associated with the selected SNPs; SOX5 rs34616559, CCDC26/GSDMC rs7833174 and DCC rs4384683. This absent or weak association suggests that the SNPs previously associated with chronic back pain are not useful as prognostic biomarkers for LBP with persistent radiculopathy.
Collapse
Affiliation(s)
- Marie Udnesseter Lie
- Research and Communication Unit for Musculoskeletal Health (FORMI), Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
- *Correspondence: Marie Udnesseter Lie,
| | - Linda Margareth Pedersen
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Physiotherapy, Oslo Metropolitan University, Oslo, Norway
| | - Ingrid Heuch
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| | - Bendik Winsvold
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Johannes Gjerstad
- Department of Work Psychology and Physiology, National Institute of Occupational Health, Oslo, Norway
- Department of Bioscience, University of Oslo, Oslo, Norway
| | - Eivind Hasvik
- Department of Physical Medicine and Rehabilitation, Østfold Hospital Trust, Grålum, Norway
| | - Øystein Petter Nygaard
- Department of Neurosurgery, St Olavs University Hospital, Trondheim, Norway
- Department of Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- National Advisory Unit on Spinal Surgery, St Olavs Hospital, Trondheim, Norway
| | - Margreth Grotle
- Research and Communication Unit for Musculoskeletal Health (FORMI), Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Physiotherapy, Oslo Metropolitan University, Oslo, Norway
| | - Dagfinn Matre
- Department of Work Psychology and Physiology, National Institute of Occupational Health, Oslo, Norway
| | - John-Anker Zwart
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
14
|
Kong H, Sun ML, Zhang XA, Wang XQ. Crosstalk Among circRNA/lncRNA, miRNA, and mRNA in Osteoarthritis. Front Cell Dev Biol 2022; 9:774370. [PMID: 34977024 PMCID: PMC8714905 DOI: 10.3389/fcell.2021.774370] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a joint disease that is pervasive in life, and the incidence and mortality of OA are increasing, causing many adverse effects on people's life. Therefore, it is very vital to identify new biomarkers and therapeutic targets in the clinical diagnosis and treatment of OA. ncRNA is a nonprotein-coding RNA that does not translate into proteins but participates in protein translation. At the RNA level, it can perform biological functions. Many studies have found that miRNA, lncRNA, and circRNA are closely related to the course of OA and play important regulatory roles in transcription, post-transcription, and post-translation, which can be used as biological targets for the prevention, diagnosis, and treatment of OA. In this review, we summarized and described the various roles of different types of miRNA, lncRNA, and circRNA in OA, the roles of different lncRNA/circRNA-miRNA-mRNA axis in OA, and the possible prospects of these ncRNAs in clinical application.
Collapse
Affiliation(s)
- Hui Kong
- College of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Ming-Li Sun
- College of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Xin-An Zhang
- College of Kinesiology, Shenyang Sport University, Shenyang, China
| | - Xue-Qiang Wang
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, China.,Department of Rehabilitation Medicine, Shanghai Shangti Orthopaedic Hospital, Shanghai, China
| |
Collapse
|
15
|
KLF4, negatively regulated by miR-7, suppresses osteoarthritis development via activating TGF-β1 signaling. Int Immunopharmacol 2021; 102:108416. [PMID: 34891002 DOI: 10.1016/j.intimp.2021.108416] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/12/2021] [Accepted: 11/25/2021] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) is a chronic degenerative disease which seriously affects the patients' daily activities and quality of life. In our previous findings, we demonstrated that overexpression of miR-7 was found in OA and promoted OA development. Its exact mechanism remains unclear. Herein, we confirmed that KLF4 was the target gene of miR-7 and KLF4 was down-regulated in human OA tissues and OA chondrocyte. KLF4 was negatively modulated by miR-7 via dual luciferase reporter assay. Cartilage-specific genes (SOX9, COL2A1, RUNX2, MMP13) are crucial regulators in cartilage degeneration. Through qRT-PCR and western blot, we observed that KLF4 overexpression could increase the expression of SOX9 and COL2A1, decrease RUNX2 and MMP13. In the meanwhile, miR-7 was proven to regulate the expression of the above cartilage-specific genes by targeting KLF4, which demonstrated KLF4 could prevent OA development. Subsequently, KLF4 also activated TGF-β1 signaling pathway, thereby affecting OA progression. Excessive KLF4 could up-regulate TGF-β1 and p-Smad2/3 level, and Smad4 level was prevented in OA chondrocytes, while adding TGF-β1 inhibitor SB525334 could rescue this impact, along with reduced TGF-β1 and p-Smad2/3 level, enriched Smad4 level. KLF4 could also reverse the effect of miR-7 on TGF-β1 signaling. Besides, it was confirmed that KLF4 could improve OA in rat OA models by HE and Safranin O-Fast green staining, and immunohistochemistry. Collectively, our findings will give more detailed evidence about miR-7 and KLF4 in OA diagnosis and treatment.
Collapse
|
16
|
Dalle Carbonare L, Bertacco J, Marchetto G, Cheri S, Deiana M, Minoia A, Tiso N, Mottes M, Valenti MT. Methylsulfonylmethane enhances MSC chondrogenic commitment and promotes pre-osteoblasts formation. Stem Cell Res Ther 2021; 12:326. [PMID: 34090529 PMCID: PMC8180127 DOI: 10.1186/s13287-021-02396-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 05/18/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Methylsulfonylmethane (MSM) is a nutraceutical compound which has been indicated to counteract osteoarthritis, a cartilage degenerative disorder. In addition, MSM has also been shown to increase osteoblast differentiation. So far, few studies have investigated MSM role in the differentiation of mesenchymal stem cells (MSCs), and no study has been performed to evaluate its overall effects on both osteogenic and chondrogenic differentiation. These two mutually regulated processes share the same progenitor cells. METHODS Therefore, with the aim to evaluate the effects of MSM on chondrogenesis and osteogenesis, we analyzed the expression of SOX9, RUNX2, and SP7 transcription factors in vitro (mesenchymal stem cells and chondrocytes cell lines) and in vivo (zebrafish model). Real-time PCR as well Western blotting, immunofluorescence, and specific in vitro and in vivo staining have been performed. Student's paired t test was used to compare the variation between the groups. RESULTS Our data demonstrated that MSM modulates the expression of differentiation-related genes both in vitro and in vivo. The increased SOX9 expression suggests that MSM promotes chondrogenesis in treated samples. In addition, RUNX2 expression was not particularly affected by MSM while SP7 expression increased in all MSM samples/model analyzed. As SP7 is required for the final commitment of progenitors to preosteoblasts, our data suggest a role of MSM in promoting preosteoblast formation. In addition, we observed a reduced expression of the osteoclast-surface receptor RANK in larvae and in scales as well as a reduced pERK/ERK ratio in fin and scale of MSM treated zebrafish. CONCLUSIONS In conclusion, our study provides new insights into MSM mode of action and suggests that MSM is a useful tool to counteract skeletal degenerative diseases by targeting MSC commitment and differentiation.
Collapse
Affiliation(s)
- Luca Dalle Carbonare
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Jessica Bertacco
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 10, 37100, Verona, Italy
| | - Giulia Marchetto
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Samuele Cheri
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Michela Deiana
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Arianna Minoia
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, I-35131, Padova, Italy
| | - Monica Mottes
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie, 10, 37100, Verona, Italy
| | - Maria Teresa Valenti
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy.
| |
Collapse
|
17
|
Zhou X, Xu W, Wang Y, Zhang H, Zhang L, Li C, Yao S, Huang Z, Huang L, Luo D. LncRNA DNM3OS regulates GREM2 via miR-127-5p to suppress early chondrogenic differentiation of rat mesenchymal stem cells under hypoxic conditions. Cell Mol Biol Lett 2021; 26:22. [PMID: 34049478 PMCID: PMC8161583 DOI: 10.1186/s11658-021-00269-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Improved chondrogenic differentiation of mesenchymal stem cells (MSCs) by genetic regulation is a potential method for regenerating articular cartilage. MiR-127-5p has been reported to promote cartilage differentiation of rat bone marrow MSCs (rMSCs); however, the regulatory mechanisms underlying hypoxia-stimulated chondrogenic differentiation remain unknown. Methods rMSCs were induced to undergo chondrogenic differentiation under normoxic or hypoxic conditions. Expression of lncRNA DNM3OS, miR-127-5p, and GREM2 was detected by quantitative real-time PCR. Proteoglycans were detected by Alcian blue staining. Western blot assays were performed to examine the relative levels of GREM2 and chondrogenic differentiation related proteins. Luciferase reporter assays were performed to assess the association among DNM3OS, miR-127-5p, and GREM2. Results MiR-127-5p levels were upregulated, while DNM3OS and GREM2 levels were downregulated in rMSCs induced to undergo chondrogenic differentiation, and those changes were attenuated by hypoxic conditions (1% O2). Further in vitro experiments revealed that downregulation of miR-127-5p reduced the production of proteoglycans and expression of chondrogenic differentiation markers (COL1A1, COL2A1, SOX9, and ACAN) and osteo/chondrogenic markers (BMP-2, p-SMAD1/2). MiR-127-5p overexpression produced the opposite results in rMSCs induced to undergo chondrogenic differentiation under hypoxic conditions. GREM2 was found to be a direct target of miR-127-5p, which was suppressed in rMSCs undergoing chondrogenic differentiation. Moreover, DNM3OS could directly bind to miR-127-5p and inhibit chondrogenic differentiation of rMSCs via regulating GREM2. Conclusions Our study revealed a novel molecular pathway (DNM3OS/miR-127-5p/GREM2) that may be involved in hypoxic chondrogenic differentiation.
Collapse
Affiliation(s)
- Xiaozhong Zhou
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wangyang Xu
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Yeyang Wang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Hui Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Li Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Chao Li
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Shun Yao
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Zixiang Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Lishan Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China
| | - Dixin Luo
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, No. 466, Xingangzhong Road, Haizhu District, Guangzhou, 510317, Guangdong, People's Republic of China.
| |
Collapse
|
18
|
Li Q, Du X, Liu L, Liu H, Pan Z, Li Q. Upregulation of miR-146b promotes porcine ovarian granulosa cell apoptosis by attenuating CYP19A1. Domest Anim Endocrinol 2021; 74:106509. [PMID: 32653739 DOI: 10.1016/j.domaniend.2020.106509] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 06/04/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are 21- to 24-nucleotide long small noncoding RNAs, which play an important role in follicular atresia and granulosa cell (GC) apoptosis in the mammalian ovary. Here, we report that miR-146b, a conserved and ovary-enriched miRNA, modulates estradiol (E2) secretion, GC apoptosis, and follicular atresia in pigs. Genome-wide analysis and quantitative real-time PCR revealed that miR-146b was significantly upregulated during follicular atresia, and fluorescence-activated cell sorting showed that miR-146b functioned as a proapoptotic factor to induce GC apoptosis. MicroRNA-mRNA network analysis and luciferase reporter assays showed that CYP19A1, the pivotal enzyme for E2 synthesis signaling, was directly targeted by miR-146b. Furthermore, miR-146b interacted with the 3'untranslated region of CYP19A1 to prevent translation, thereby regulating CYP19A1-mediated E2 secretion and GC apoptosis. However, miR-146b was not regulated by the transcription factor SMAD4 or oxidative stress, both of which are critical regulators of CYP19A1. We, thus, conclude that miR-146b is a novel epigenetic factor regulating GC functions, follicular development, and female reproduction.
Collapse
Affiliation(s)
- Q Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - X Du
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - L Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - H Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Z Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Q Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
19
|
De Luna A, Otahal A, Nehrer S. Mesenchymal Stromal Cell-Derived Extracellular Vesicles - Silver Linings for Cartilage Regeneration? Front Cell Dev Biol 2020; 8:593386. [PMID: 33363147 PMCID: PMC7758223 DOI: 10.3389/fcell.2020.593386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/19/2020] [Indexed: 01/15/2023] Open
Abstract
As the world's population is aging, the incidence of the degenerative disease Osteoarthritis (OA) is increasing. Current treatment options of OA focus on the alleviation of the symptoms including pain and inflammation rather than on restoration of the articular cartilage. Cell-based therapies including the application of mesenchymal stromal cells (MSCs) have been a promising tool for cartilage regeneration approaches. Due to their immunomodulatory properties, their differentiation potential into cells of the mesodermal lineage as well as the plurality of sources from which they can be isolated, MSCs have been applied in a vast number of studies focusing on the establishment of new treatment options for Osteoarthritis. Despite promising outcomes in vitro and in vivo, applications of MSCs are connected with teratoma formation, limited lifespan of differentiated cells as well as rejection of the cells after transplantation, highlighting the need for new cell free approaches harboring the beneficial properties of MSCs. It has been demonstrated that the regenerative potential of MSCs is mediated by the release of paracrine factors rather than by differentiation into cells of the desired tissue. Besides soluble factors, extracellular vesicles are the major component of a cell's secretome. They represent novel mechanisms by which (pathogenic) signals can be communicated between cell types as they deliver bioactive molecules (nucleic acids, proteins, lipids) from the cell of origin to the target cell leading to specific biological processes upon uptake. This review will give an overview about extracellular vesicles including general characteristics, isolation methods and characterization approaches. Furthermore, the role of MSC-derived extracellular vesicles in in vitro and in vivo studies for cartilage regeneration will be summarized with special focus on transported miRNA which either favored the progression of OA or protected the cartilage from degradation. In addition, studies will be reviewed investigating the impact of MSC-derived extracellular vesicles on inflammatory arthritis. As extracellular vesicles are present in all body fluids, their application as potential biomarkers for OA will also be discussed in this review. Finally, studies exploring the combination of MSC-derived extracellular vesicles with biomaterials for tissue engineering approaches are summarized.
Collapse
Affiliation(s)
- Andrea De Luna
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | | | | |
Collapse
|
20
|
Witte K, de Andrés MC, Wells J, Dalby MJ, Salmeron-Sanchez M, Oreffo ROC. Chondrobags: A high throughput alginate-fibronectin micromass platform for in vitro human cartilage formation. Biofabrication 2020; 12:045034. [DOI: 10.1088/1758-5090/abb653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
21
|
Yan Y, Chang C, Su J, Venø MT, Kjems J. Osteoblastogenesis Alters Small RNA Profiles in EVs Derived from Bone Marrow Stem Cells (BMSCs) and Adipose Stem Cells (ASCs). Biomedicines 2020; 8:biomedicines8100387. [PMID: 32998458 PMCID: PMC7599808 DOI: 10.3390/biomedicines8100387] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
Multipotent stem cells (MSCs) are used in various therapeutic applications based on their paracrine secretion activity. Here, we set out to identify and characterize the paracrine factors released during osteoblastogenesis, with a special focus on small non-coding RNAs released in extracellular vesicles (EVs). Bone marrow stem cells (BMSCs) and adipose stem cells (ASCs) from healthy human donors were used as representatives of MSCs. We isolated EVs secreted before and after induction of osteoblastic differentiation and found that the EVs contained a specific subset of microRNAs (miRNAs) and tRNA-derived small RNAs (tsRNA) compared to their parental cells. Osteoblastic differentiation had a larger effect on the small RNA profile of BMSC-EVs relative to ASC-EVs. Our data showed that EVs from different MSC origin exhibited distinct expression profiles of small RNA profiles when undergoing osteoblastogenesis, a factor that should be taken into consideration for stem cell therapy.
Collapse
Affiliation(s)
- Yan Yan
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (C.C.); (J.S.); (M.T.V.)
- Omiics ApS, Åbogade 15, 8200 Aarhus N, Denmark
| | - Clare Chang
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (C.C.); (J.S.); (M.T.V.)
| | - Junyi Su
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (C.C.); (J.S.); (M.T.V.)
| | - Morten T. Venø
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (C.C.); (J.S.); (M.T.V.)
- Omiics ApS, Åbogade 15, 8200 Aarhus N, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (C.C.); (J.S.); (M.T.V.)
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
- Correspondence: ; Tel.: +45-289-920-86
| |
Collapse
|
22
|
Xie F, Liu YL, Chen XY, Li Q, Zhong J, Dai BY, Shao XF, Wu GB. Role of MicroRNA, LncRNA, and Exosomes in the Progression of Osteoarthritis: A Review of Recent Literature. Orthop Surg 2020; 12:708-716. [PMID: 32436304 PMCID: PMC7307224 DOI: 10.1111/os.12690] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a common clinical degenerative disease characterized by the destruction of articular cartilage, which has an increasing impact on people's lives and social economy. The pathogenesis of OA is complex and unclear, and there is no effective way to block its progress. The study of the pathogenesis of OA is the prerequisite for the early diagnosis and effective treatment of OA. To define the pathogenesis of OA, this review considers the pathological mechanism of OA that involves microRNA, lncRNA, and exosomes. More and more evidence shows that microRNA, lncRNA, and exosomes are closely related to OA. MicroRNA inhibits the target gene by binding to the 3'- untranslated region of the targets. LncRNA usually competes with microRNA to regulate the expression level of downstream genes, while exosomes, as a carrier of intercellular information transfer, transmit the biological information of mother cells to target cells, and the effect of exosomes secreted by different cells on OA are different. In this review, we emphasized that different microRNA, lncRNA, and exosomes have different regulatory effects on chondrocyte proliferation and apoptosis, extracellular matrix degradation and inflammation. Besides, we classified and analyzed these molecules according to their effects on the progress of OA. Based on the analysis of the reported literature, this review reveals some pathogenesis of OA, and emphasizes that microRNA, lncRNA, and exosomes have great potential to assist early diagnosis and effective treatment of OA.
Collapse
Affiliation(s)
- Fang Xie
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Yong-Li Liu
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Xiu-Yuan Chen
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Qian Li
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Jia Zhong
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Bin-Yu Dai
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Xian-Fang Shao
- Affiliated Changde Hospital, Hunan University of Traditional Chinese Medicine, Changde, China
| | - Guan-Bao Wu
- Department of Orthopaedics, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
| |
Collapse
|
23
|
Barreto G, Manninen M, K. Eklund K. Osteoarthritis and Toll-Like Receptors: When Innate Immunity Meets Chondrocyte Apoptosis. BIOLOGY 2020; 9:biology9040065. [PMID: 32235418 PMCID: PMC7235883 DOI: 10.3390/biology9040065] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/26/2020] [Accepted: 03/28/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) has long been viewed as a degenerative disease of cartilage, but accumulating evidence indicates that inflammation has a critical role in its pathogenesis. In particular, chondrocyte-mediated inflammatory responses triggered by the activation of innate immune receptors by alarmins (also known as danger signals) are thought to be involved. Thus, toll-like receptors (TLRs) and their signaling pathways are of particular interest. Recent reports suggest that among the TLR-induced innate immune responses, apoptosis is one of the critical events. Apoptosis is of particular importance, given that chondrocyte death is a dominant feature in OA. This review focuses on the role of TLR signaling in chondrocytes and the role of TLR activation in chondrocyte apoptosis. The functional relevance of TLR and TLR-triggered apoptosis in OA are discussed as well as their relevance as candidates for novel disease-modifying OA drugs (DMOADs).
Collapse
Affiliation(s)
- Goncalo Barreto
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, 00014 Helsinki, Finland;
- Translational Immunology Research Program, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-4585-381-10
| | | | - Kari K. Eklund
- Department of Rheumatology, Helsinki University and Helsinki University Hospital, 00014 Helsinki, Finland;
- Translational Immunology Research Program, University of Helsinki, 00014 Helsinki, Finland
- Orton Research Institute, 00280 Helsinki, Finland;
| |
Collapse
|
24
|
COL2A1 and Caspase-3 as Promising Biomarkers for Osteoarthritis Prognosis in an Equus asinus Model. Biomolecules 2020; 10:biom10030354. [PMID: 32111016 PMCID: PMC7175237 DOI: 10.3390/biom10030354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis (OA) is one of the most degenerative joint diseases in both human and veterinary medicine. The objective of the present study was the early diagnosis of OA in donkeys using a reliable grading of the disease based on clinical, chemical, and molecular alterations. OA was induced by intra-articular injection of 25 mg monoiodoacetate (MIA) as a single dose into the left radiocarpal joint of nine donkeys. Animals were clinically evaluated through the assessment of lameness score, radiographic, and ultrasonographic findings for seven months. Synovial fluid and cartilage samples were collected from both normal and diseased joints for the assessment of matrix metalloproteinases (MMPs) activity, COL2A1 protein expression level, and histopathological and immunohistochemical analysis of Caspase-3. Animals showed the highest lameness score post-induction after one week then decreased gradually with the progression of radiographical and ultrasonographic changes. MMP activity and COL2A1 and Caspase-3 expression increased, accompanied by articular cartilage degeneration and loss of proteoglycan. OA was successfully graded in Egyptian donkeys, with the promising use of COL2A1and Caspase-3 for prognosis. However, MMPs failed to discriminate between early and late grades of OA.
Collapse
|
25
|
Yan X, Chen YR, Song YF, Yang M, Ye J, Zhou G, Yu JK. Scaffold-Based Gene Therapeutics for Osteochondral Tissue Engineering. Front Pharmacol 2020; 10:1534. [PMID: 31992984 PMCID: PMC6970981 DOI: 10.3389/fphar.2019.01534] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 11/27/2019] [Indexed: 12/20/2022] Open
Abstract
Significant progress in osteochondral tissue engineering has been made for biomaterials designed to deliver growth factors that promote tissue regeneration. However, due to diffusion characteristics of hydrogels, the accurate delivery of signaling molecules remains a challenge. In comparison to the direct delivery of growth factors, gene therapy can overcome these challenges by allowing the simultaneous delivery of growth factors and transcription factors, thereby enhancing the multifactorial processes of tissue formation. Scaffold-based gene therapy provides a promising approach for tissue engineering through transfecting cells to enhance the sustained expression of the protein of interest or through silencing target genes associated with bone and joint disease. Reports of the efficacy of gene therapy to regenerate bone/cartilage tissue regeneration are widespread, but reviews on osteochondral tissue engineering using scaffold-based gene therapy are sparse. Herein, we review the recent advances in gene therapy with a focus on tissue engineering scaffolds for osteochondral regeneration.
Collapse
Affiliation(s)
- Xin Yan
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - You-Rong Chen
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Yi-Fan Song
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Meng Yang
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Jing Ye
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| | - Gang Zhou
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jia-Kuo Yu
- Knee Surgery Department of the Institute of Sports Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
26
|
Cheng F, Hu H, Sun K, Yan F, Geng Y. miR-455-3p enhances chondrocytes apoptosis and inflammation by targeting COL2A1 in the in vitro osteoarthritis model. Biosci Biotechnol Biochem 2019; 84:695-702. [PMID: 31809639 DOI: 10.1080/09168451.2019.1690974] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Emerging evidence has shown that microRNAs are important regulators in osteoarthritis (OA). Here, we investigated the function role of miR-455-3p in the pathogenesis of OA and the underlying molecular mechanisms. We first established the in vitro OA model using IL-1β treated human chondrocyte cell line CHON-001. Using quantitative real time PCR, we observed the expression of miR-455-3p expression was up-regulated in the OA cartilage tissues and IL-1β-treated chondrocytes. A series of function assays, including CCK-8 assay, flow cytometry, and ELISA assay showed that miR-455-3p contributed to IL-1β-induced apoptosis and inflammation. Moreover, COL2A1 was confirmed as a target of miR-455-3p by luciferase reporter assay. Furthermore, COL2A1 knockdown reversed the effects of miR-455-3p inhibition, and aggravated the effects of miR-455-3p overexpression on IL-1β-induced OA-like phenomenon. Taken together, these results revealed that miR-455-3p/COL2A1 axis might provide a novel molecular target for the treatment of OA.
Collapse
Affiliation(s)
- Fang Cheng
- Department of Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Haiyan Hu
- Department of Traditional Chinese and Western Medicine, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Kefu Sun
- Department of Orthopedic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Fengfeng Yan
- Department of Pain Clinic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| | - Yuqiang Geng
- Department of Orthopedic, The Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, China
| |
Collapse
|
27
|
Wang Y, Wu C, Yang Y, Ren Z, Lammi MJ, Guo X. Preliminary Exploration of hsa_circ_0032131 Levels in Peripheral Blood as a Potential Diagnostic Biomarker of Osteoarthritis. Genet Test Mol Biomarkers 2019; 23:717-721. [PMID: 31526191 DOI: 10.1089/gtmb.2019.0036] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Osteoarthritis (OA) is a common chronic degenerative joint disease characterized by articular cartilage degeneration and synovitis. CircRNAs are increasingly being recognized as functional endogenous RNAs with a stable structure and high tissue specificity. Recent studies have shown that some circRNAs may be involved in the initiation and progression of OA and that there is differential expression of circRNAs in chondrocytes in vitro isolated from patients with OA. Purpose: In this study, we aimed to determine if circRNA levels in the peripheral blood of Chinese Han patients with OA would be diagnostic based on the previous in vitro studies. Methods: We collected peripheral blood samples from 25 patients suffering from OA and 25 healthy controls and measured hsa_circ_0032131_CBC1 RNA levels through quantitative RT-PCR (qRT-PCR). The statistical basis for evaluating the diagnostic value was to calculate the area under the receiver operator characteristic (ROC) curve. Results: The results of the qRT-PCR for hsa_circ_0032131_CBC1 were consistent with those of the microarray analysis. The ROC curve shows that hsa_circ_0032131 holds diagnostic value for OA (0.8455, p < 0.01). Conclusions: Our research indicates that differentially expressed circRNAs may be involved in the development of OA and could be used diagnostically.
Collapse
Affiliation(s)
- Ying Wang
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, People's Republic of China
| | - Cuiyan Wu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an, Shaanxi, People's Republic of China
| | - Yimin Yang
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, People's Republic of China
| | - Zhiwei Ren
- Department of Orthopedics, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi, People's Republic of China
| | - Mikko J Lammi
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an, Shaanxi, People's Republic of China.,Department of Integrative Medical Biology, University of Umeå, Umeå, Sweden
| | - Xiong Guo
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
28
|
Liu X, Liu L, Zhang H, Shao Y, Chen Z, Feng X, Fang H, Zhao C, Pan J, Zhang H, Zeng C, Cai D. MiR-146b accelerates osteoarthritis progression by targeting alpha-2-macroglobulin. Aging (Albany NY) 2019; 11:6014-6028. [PMID: 31422941 PMCID: PMC6738400 DOI: 10.18632/aging.102160] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/05/2019] [Indexed: 01/14/2023]
Abstract
Osteoarthritis (OA) is an aging-related chronic degenerative disease characterized by the degradation of chondrocyte extracellular matrix (ECM). Previous studies have suggested that microRNAs (miRNAs) are associated with OA, but the role of miR-146b in OA remains unclear. The aim of this study was to determine the role of miR-146b in OA progression. The effect of miR-146b on ECM degradation were studied in mouse chondrocytes transfected with miRNA and treated with IL-1β. Cell viability and the expression levels of proteolytic enzymes in the transfected cells were assessed by real-time RT-PCR, ELISA and Western blots. We found downregulation of miR-146b expression in chondrocytes dramatically inhibited IL-1β-induced caspase activation and proteolytic enzyme expression via influencing its targeted Alpha-2-macroglobulin (A2M). Luciferase reporter assays confirmed that A2M mRNA was negatively regulated by miR-146b in chondrocytes. Intra-articular injection of antago-miR-146b against miR-146b effectively protected mice from the progression of DMM-induced osteoarthritis by inhibiting cartilage proteoglycan degradation. Our study indicates that miR-146b plays a critical role in the progression of injury-induced osteoarthritis by directly targeting A2M expression to elevate the proteolytic enzyme production and stimulate chondrocytes apoptosis, and miR-146b as well as A2M could be therapeutic targets.
Collapse
Affiliation(s)
- Xin Liu
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Liangliang Liu
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Hongbo Zhang
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Yan Shao
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Ziyu Chen
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Xiaofeng Feng
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Hang Fang
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Chang Zhao
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Jianying Pan
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Haiyan Zhang
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Chun Zeng
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| | - Daozhang Cai
- Department of Orthopedics, Academy of Orthopaedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
- Orthopaedic Hospital of Guangdong Province, Guangzhou 510630, China
| |
Collapse
|
29
|
Mechanosensitive MiRs regulated by anabolic and catabolic loading of human cartilage. Osteoarthritis Cartilage 2019; 27:1208-1218. [PMID: 31009748 DOI: 10.1016/j.joca.2019.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 02/26/2019] [Accepted: 04/10/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Elucidation of whether miRs are involved in mechanotransduction pathways by which cartilage is maintained or disturbed has a particular importance in our understanding of osteoarthritis (OA) pathophysiology. The aim was to investigate whether mechanical loading influences global miR-expression in human chondrocytes and to identify mechanosensitive miRs responding to beneficial and non-beneficial loading regimes as potential to obtain valuable diagnostic or therapeutic targets to advance OA-treatment. METHOD Mature tissue-engineered human cartilage was subjected to two distinct loading regimes either stimulating or suppressing proteoglycan-synthesis, before global miR microarray analysis. Promising candidate miRs were selected, re-evaluated by qRT-PCR and tested for expression in human healthy vs OA cartilage samples. RESULTS After anabolic loading, miR microarray profiling revealed minor changes in miR-expression while catabolic stimulation produced a significant regulation of 80 miRs with a clear separation of control and compressed samples by hierarchical clustering. Cross-testing of selected miRs revealed that miR-221, miR-6872-3p, miR-6723-5p were upregulated by both loading conditions while others (miR-199b-5p, miR-1229-5p, miR-1275, miR-4459, miR-6891-5p, miR-7150) responded specifically after catabolic loading. Mechanosensitivity of miR-221 correlated with pERK1/2-activation induced by both loading conditions. The miR-response to loading was transient and a constitutive deregulation of mechano-miRs in OA vs healthy articular cartilage was not observed. CONCLUSIONS MiRs with broader vs narrower mechanosensitivity were discovered and the first group of mechanosensitive miRs characteristic for non-beneficial loading was defined that may shape the proteome differentially when cartilage tissue is disturbed. The findings prompt future investigations into miR-relevance for mechano-responsive pathways and the corresponding miR-target molecules.
Collapse
|
30
|
Costa Moura C, Lanham SA, Monfort T, Bourdakos KN, Tare RS, Oreffo ROC, Mahajan S. Quantitative temporal interrogation in 3D of bioengineered human cartilage using multimodal label-free imaging. Integr Biol (Camb) 2019; 10:635-645. [PMID: 30225469 DOI: 10.1039/c8ib00050f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The unique properties of skeletal stem cells have attracted significant attention in the development of strategies for skeletal regeneration. However, there remains a crucial unmet need to develop quantitative tools to elucidate skeletal cell development and monitor the formation of regenerated tissues using non-destructive techniques in 3D. Label-free methods such as coherent anti-Stokes Raman scattering (CARS), second harmonic generation (SHG) and two-photon excited auto-fluorescence (TPEAF) microscopy are minimally invasive, non-destructive, and present new powerful alternatives to conventional imaging techniques. Here we report a combination of these techniques in a single multimodal system for the temporal assessment of cartilage formation by human skeletal cells. The evaluation of bioengineered cartilage, with a new parameter measuring the amount of collagen per cell, collagen fibre structure and chondrocyte distribution, was performed using the 3D non-destructive platform. Such 3D label-free temporal quantification paves the way for tracking skeletal cell development in real-time and offers a paradigm shift in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Catarina Costa Moura
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK. and Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| | - Stuart A Lanham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| | - Tual Monfort
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| | - Konstantinos N Bourdakos
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| | - Rahul S Tare
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK. and Mechanical Engineering Department, Faculty of Engineering and the Environment, University of Southampton, Highfield Campus, Southampton, SO17 1BJ, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK.
| | - Sumeet Mahajan
- Department of Chemistry and Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.
| |
Collapse
|
31
|
Wang P, Dong R, Wang B, Lou Z, Ying J, Xia C, Hu S, Wang W, Sun Q, Zhang P, Ge Q, Xiao L, Chen D, Tong P, Li J, Jin H. Genome-wide microRNA screening reveals miR-582-5p as a mesenchymal stem cell-specific microRNA in subchondral bone of the human knee joint. J Cell Physiol 2019; 234:21877-21888. [PMID: 31049977 PMCID: PMC6767428 DOI: 10.1002/jcp.28751] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 12/22/2022]
Abstract
Emerging evidence suggests that microRNAs (miRNAs) may be pathologically involved in osteoarthritis (OA). Subchondral bone (SCB) sclerosis is accounted for the knee osteoarthritis (KOA) development and progression. In this study, we aimed to screen the miRNA biomarkers of KOA and investigated whether these miRNAs regulate the differentiation potential of mesenchymal stem cells (MSCs) and thus contributing to SCB. We identified 48 miRNAs in the blood samples in KOA patients (n = 5) through microarray expression profiling detection. After validation with larger sample number, we confirmed hsa-miR-582-5p and hsa-miR-424-5p were associated with the pathology of SCB sclerosis. Target genes prediction and pathway analysis were implemented with online databases, indicating these two candidate miRNAs were closely related to the pathways of pluripotency of stem cells and pathology of OA. Surprisingly, mmu-miR-582-5p (homology of hsa-miR-582-5p) was downregulated in osteogenic differentiation and upregulated in adipogenic differentiation of mesenchymal progenitor C3H10T1/2 cells, whereas mmu-mir-322-5p (homology of hsa-miR-424-5p) showed no change through the in vitro study. Supplementing mmu-miR-582-5p mimics blocked osteogenic and induced adipogenic differentiation of C3H10T1/2 cells, whereas silencing of the endogenous mmu-miR-582-5p enhanced osteogenic and repressed adipogenic differentiation. Further mechanism studies showed that mmu-miR-582-5p was directly targeted to Runx2. Mutation of putative mmu-miR-582-5p binding sites in Runx2 3' untranslated region (3'UTR) could abolish the response of the 3'UTR-luciferase construct to mmu-miR-582-5p supplementation. Generally speaking, our data suggest that miR-582-5p is an important biomarker of KOA and is able to regulate osteogenic and adipogenic differentiation of MSCs via targeting Runx2. The study also suggests that miR-582-5p may play a crucial role in SCB sclerosis of human KOA.
Collapse
Affiliation(s)
- Pinger Wang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Institute of Orthopaedic and Traumatology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Rui Dong
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Institute of Orthopaedic and Traumatology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Baoli Wang
- Key Laboratory of Hormones and Development, Ministry of Health, Tianjin Metabolic Diseases Hospital, Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Zhaohuan Lou
- The Pharmaceutical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jun Ying
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Chenjie Xia
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Songfeng Hu
- Department of Orthopaedics, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing, Zhejiang, China
| | - Weidong Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qi Sun
- Department of Orthopaedic Surgery, Fuyang Orthopaedics and Traumatology Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Peng Zhang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qinwen Ge
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Luwei Xiao
- Institute of Orthopaedic and Traumatology of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, Illinois
| | - Peijian Tong
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ju Li
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hongting Jin
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.,Institute of Orthopaedic and Traumatology of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Abstract
SOX transcription factors participate in the specification, differentiation and activities of many cell types in development and beyond. The 20 mammalian family members are distributed into eight groups based on sequence identity, and while co-expressed same-group proteins often have redundant functions, different-group proteins typically have distinct functions. More than a handful of SOX proteins have pivotal roles in skeletogenesis. Heterozygous mutations in their genes cause human diseases, in which skeletal dysmorphism is a major feature, such as campomelic dysplasia (SOX9), or a minor feature, such as LAMSHF syndrome (SOX5) and Coffin-Siris-like syndromes (SOX4 and SOX11). Loss- and gain-of-function experiments in animal models have revealed that SOX4 and SOX11 (SOXC group) promote skeletal progenitor survival and control skeleton patterning and growth; SOX8 (SOXE group) delays the differentiation of osteoblast progenitors; SOX9 (SOXE group) is essential for chondrocyte fate maintenance and differentiation, and works in cooperation with SOX5 and SOX6 (SOXD group) and other types of transcription factors. These and other SOX proteins have also been proposed, mainly through in vitro experiments, to have key roles in other aspects of skeletogenesis, such as SOX2 in osteoblast stem cell self-renewal. We here review current knowledge of well-established and proposed skeletogenic roles of SOX proteins, their transcriptional and non-transcriptional actions, and their modes of regulation at the gene, RNA and protein levels. We also discuss gaps in knowledge and directions for future research to further decipher mechanisms underlying skeletogenesis in health and diseases and identify treatment options for skeletal malformation and degeneration diseases.
Collapse
Affiliation(s)
- Véronique Lefebvre
- The Children's Hospital of Philadelphia, Philadelphia, PA, United States.
| |
Collapse
|
33
|
Lolli A, Colella F, De Bari C, van Osch GJVM. Targeting anti-chondrogenic factors for the stimulation of chondrogenesis: A new paradigm in cartilage repair. J Orthop Res 2019; 37:12-22. [PMID: 30175861 DOI: 10.1002/jor.24136] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/09/2018] [Indexed: 02/04/2023]
Abstract
Trauma and age-related cartilage disorders represent a major global cause of morbidity, resulting in chronic pain and disability in patients. A lack of effective therapies, together with a rapidly aging population, creates an impressive clinical and economic burden on healthcare systems. In this scenario, experimental therapies based on transplantation or in situ stimulation of skeletal Mesenchymal Stem/progenitor Cells (MSCs) have raised great interest for cartilage repair. Nevertheless, the challenge of guiding MSC differentiation and preventing cartilage hypertrophy and calcification still needs to be overcome. While research has mostly focused on the stimulation of cartilage anabolism using growth factors, several issues remain unresolved prompting the field to search for novel solutions. Recently, inhibition of anti-chondrogenic regulators has emerged as an intriguing opportunity. Anti-chondrogenic regulators include extracellular proteins as well as intracellular transcription factors and microRNAs that act as potent inhibitors of pro-chondrogenic signals. Suppression of these inhibitors can enhance MSC chondrogenesis and production of cartilage matrix. We here review the current knowledge concerning different types of anti-chondrogenic regulators. We aim to highlight novel therapeutic targets for cartilage repair and discuss suitable tools for suppressing their anti-chondrogenic functions. Further effort is needed to unveil the therapeutic perspectives of this approach and pave the way for effective treatment of cartilage injuries in patients. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands
| | - Fabio Colella
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Wytemaweg 80, 3015CN Rotterdam, the Netherlands.,Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
34
|
Cheng VKF, Au PCM, Tan KC, Cheung CL. MicroRNA and Human Bone Health. JBMR Plus 2018; 3:2-13. [PMID: 30680358 PMCID: PMC6339549 DOI: 10.1002/jbm4.10115] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/05/2018] [Accepted: 10/09/2018] [Indexed: 12/19/2022] Open
Abstract
The small non‐coding microRNAs (miRNAs) are post‐transcription regulators that modulate diverse cellular process in bone cells. Because optimal miRNA targeting is essential for their function, single‐nucleotide polymorphisms (SNPs) within or proximal to the loci of miRNA (miR‐SNPs) or mRNA (PolymiRTS) could potentially disrupt the miRNA‐mRNA interaction, leading to changes in bone metabolism and osteoporosis. Recent human studies of skeletal traits using miRNA profiling, genomewide association studies, and functional studies started to decipher the complex miRNA regulatory network. These studies have indicated that miRNAs may be a promising bone marker. This review focuses on human miRNA studies on bone traits and discusses how genetic variants affect bone metabolic pathways. Major ex vivo investigations using human samples supported with animal and in vitro models have shed light on the mechanistic role of miRNAs. Furthermore, studying the miRNAs’ signatures in secondary osteoporosis and osteoporotic medications such as teriparatide (TPTD) and denosumab (DMab) have provided valuable insight into clinical management of the disease. © 2018 The Authors. JBMR Plus Published by Wiley Periodicals, Inc. on behalf of the American Society for Bone and Mineral Research
Collapse
Affiliation(s)
- Vincent Ka-Fai Cheng
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong
| | - Philip Chun-Ming Au
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong
| | - Kathryn Cb Tan
- Department of Medicine The University of Hong Kong Pokfulam Hong Kong
| | - Ching-Lung Cheung
- Department of Pharmacology and Pharmacy The University of Hong Kong Pokfulam Hong Kong.,Centre for Genomic Sciences Li Ka Shing Faculty of Medicine The University of Hong Kong Pokfulam Hong Kong
| |
Collapse
|
35
|
|
36
|
Suri P, Palmer MR, Tsepilov YA, Freidin MB, Boer CG, Yau MS, Evans DS, Gelemanovic A, Bartz TM, Nethander M, Arbeeva L, Karssen L, Neogi T, Campbell A, Mellstrom D, Ohlsson C, Marshall LM, Orwoll E, Uitterlinden A, Rotter JI, Lauc G, Psaty BM, Karlsson MK, Lane NE, Jarvik GP, Polasek O, Hochberg M, Jordan JM, Van Meurs JBJ, Jackson R, Nielson CM, Mitchell BD, Smith BH, Hayward C, Smith NL, Aulchenko YS, Williams FMK. Genome-wide meta-analysis of 158,000 individuals of European ancestry identifies three loci associated with chronic back pain. PLoS Genet 2018; 14:e1007601. [PMID: 30261039 PMCID: PMC6159857 DOI: 10.1371/journal.pgen.1007601] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 08/02/2018] [Indexed: 01/07/2023] Open
Abstract
Back pain is the #1 cause of years lived with disability worldwide, yet surprisingly little is known regarding the biology underlying this symptom. We conducted a genome-wide association study (GWAS) meta-analysis of chronic back pain (CBP). Adults of European ancestry were included from 15 cohorts in the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium, and from the UK Biobank interim data release. CBP cases were defined as those reporting back pain present for ≥3-6 months; non-cases were included as comparisons ("controls"). Each cohort conducted genotyping using commercially available arrays followed by imputation. GWAS used logistic regression models with additive genetic effects, adjusting for age, sex, study-specific covariates, and population substructure. The threshold for genome-wide significance in the fixed-effect inverse-variance weighted meta-analysis was p<5×10(-8). Suggestive (p<5×10(-7)) and genome-wide significant (p<5×10(-8)) variants were carried forward for replication or further investigation in the remaining UK Biobank participants not included in the discovery sample. The discovery sample comprised 158,025 individuals, including 29,531 CBP cases. A genome-wide significant association was found for the intronic variant rs12310519 in SOX5 (OR 1.08, p = 7.2×10(-10)). This was subsequently replicated in 283,752 UK Biobank participants not included in the discovery sample, including 50,915 cases (OR 1.06, p = 5.3×10(-11)), and exceeded genome-wide significance in joint meta-analysis (OR 1.07, p = 4.5×10(-19)). We found suggestive associations at three other loci in the discovery sample, two of which exceeded genome-wide significance in joint meta-analysis: an intergenic variant, rs7833174, located between CCDC26 and GSDMC (OR 1.05, p = 4.4×10(-13)), and an intronic variant, rs4384683, in DCC (OR 0.97, p = 2.4×10(-10)). In this first reported meta-analysis of GWAS for CBP, we identified and replicated a genetic locus associated with CBP (SOX5). We also identified 2 other loci that reached genome-wide significance in a 2-stage joint meta-analysis (CCDC26/GSDMC and DCC).
Collapse
Affiliation(s)
- Pradeep Suri
- Seattle Epidemiologic Research and Information Center (ERIC), Department of Veterans Affairs Office of Research and Development, Seattle, Washington, United States of America
- Division of Rehabilitation Care Services, VA Puget Sound Health Care System, Seattle, Washington, United States of America
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, United States of America
| | - Melody R. Palmer
- Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Yakov A. Tsepilov
- Polyomica, ‘s-Hertogenbosch, the Netherlands
- Laboratory of Theoretical and Applied Functional Genomics, Novosibirsk State University, Novosibirsk, Russia
- Laboratory of Recombination and Segregation Analysis, Institute of Cytology and Genetics SD RAS, Novosibirsk, Russia
| | - Maxim B. Freidin
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| | - Cindy G. Boer
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Michelle S. Yau
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, United States of America
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, California, United States of America
| | - Andrea Gelemanovic
- Department of Public Health, University of Split Medical School, Split, Croatia
| | - Traci M. Bartz
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Maria Nethander
- Department of Medicine, University of Göteborg, Göteborg, Sweden
| | - Liubov Arbeeva
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | | | - Tuhina Neogi
- Clinical Epidemiology Unit, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Dan Mellstrom
- Geriatric Medicine, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Sweden
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Göteborg, Sweden
| | - Lynn M. Marshall
- Department of Orthopedics and Rehabilitation, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Eric Orwoll
- Department of Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Andre Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Jerome I. Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, California, United States of America
- Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - Gordan Lauc
- Genos Ltd, Osijek, Croatia
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit and Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Health Services, University of Washington, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, United States of America
| | - Magnus K. Karlsson
- Department of Orthopedics, Skane University Hospital, Lund University, Malmö, Sweden
| | - Nancy E. Lane
- Departments of Medicine and Rheumatology, University of California Davis, Sacramento, California, United States of America
| | - Gail P. Jarvik
- Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Ozren Polasek
- Department of Public Health, University of Split Medical School, Split, Croatia
- Hospital “Sveti Ivan”, Zagreb, Croatia
| | - Marc Hochberg
- Departments of Medicine and Epidemiology, University of Maryland, Baltimore, Maryland, United States of America
| | - Joanne M. Jordan
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | | | - Rebecca Jackson
- Department of Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Carrie M. Nielson
- School of Public Health, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Braxton D. Mitchell
- Departments of Medicine and Epidemiology, University of Maryland, Baltimore, Maryland, United States of America
- Geriatric Research, Education and Clinical Center, Veterans Affairs Medical Center, Baltimore, Maryland, United States of America
| | - Blair H. Smith
- Division of Population Health Sciences, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, United Kingdom
| | - Nicholas L. Smith
- Seattle Epidemiologic Research and Information Center (ERIC), Department of Veterans Affairs Office of Research and Development, Seattle, Washington, United States of America
- Department of Epidemiology, University of Washington, Seattle, Washington, United States of America
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, United States of America
| | | | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, King’s College London, London, United Kingdom
| |
Collapse
|
37
|
Zhao W, Shen G, Ren H, Liang D, Yu X, Zhang Z, Huang J, Qiu T, Tang J, Shang Q, Yu P, Wu Z, Jiang X. Therapeutic potential of microRNAs in osteoporosis function by regulating the biology of cells related to bone homeostasis. J Cell Physiol 2018; 233:9191-9208. [PMID: 30078225 DOI: 10.1002/jcp.26939] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are novel regulatory factors that play important roles in numerous cellular processes through the posttranscriptional regulation of gene expression. Recently, deregulation of the miRNA-mediated mechanism has emerged as an important pathological factor in osteoporosis. However, a detailed molecular mechanism between miRNAs and osteoporosis is still not available. In this review, the roles of miRNAs in the regulation of cells related to bone homeostasis as well as miRNAs that deregulate in human or animal are discussed. Moreover, the miRNAs that act as clusters in the biology of cells in the bone microenvironment and the difference of some important miRNAs for bone homeostasis between bone and other organs are mentioned. Overall, miRNAs that contribute to the pathogenesis of osteoporosis and their therapeutic potential are considered.
Collapse
Affiliation(s)
- Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Gengyang Shen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinjing Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Qiu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingjing Tang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiyuan Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zixian Wu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Laboratory Affiliated to National Key Discipline of Orthopaedic and Traumatology of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
38
|
Onset and Progression of Human Osteoarthritis-Can Growth Factors, Inflammatory Cytokines, or Differential miRNA Expression Concomitantly Induce Proliferation, ECM Degradation, and Inflammation in Articular Cartilage? Int J Mol Sci 2018; 19:ijms19082282. [PMID: 30081513 PMCID: PMC6121276 DOI: 10.3390/ijms19082282] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/22/2018] [Accepted: 08/01/2018] [Indexed: 12/30/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative whole joint disease, for which no preventative or therapeutic biological interventions are available. This is likely due to the fact that OA pathogenesis includes several signaling pathways, whose interactions remain unclear, especially at disease onset. Early OA is characterized by three key events: a rarely considered early phase of proliferation of cartilage-resident cells, in contrast to well-established increased synthesis, and degradation of extracellular matrix components and inflammation, associated with OA progression. We focused on the question, which of these key events are regulated by growth factors, inflammatory cytokines, and/or miRNA abundance. Collectively, we elucidated a specific sequence of the OA key events that are described best as a very early phase of proliferation of human articular cartilage (AC) cells and concomitant anabolic/catabolic effects that are accompanied by incipient pro-inflammatory effects. Many of the reviewed factors appeared able to induce one or two key events. Only one factor, fibroblast growth factor 2 (FGF2), is capable of concomitantly inducing all key events. Moreover, AC cell proliferation cannot be induced and, in fact, is suppressed by inflammatory signaling, suggesting that inflammatory signaling cannot be the sole inductor of all early OA key events, especially at disease onset.
Collapse
|
39
|
Malemud CJ. MicroRNAs and Osteoarthritis. Cells 2018; 7:cells7080092. [PMID: 30071609 PMCID: PMC6115911 DOI: 10.3390/cells7080092] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/23/2018] [Accepted: 07/29/2018] [Indexed: 12/14/2022] Open
Abstract
An imbalance in gene expressional events skewing chondrocyte anabolic and catabolic pathways toward the latter causes an aberrant turnover and loss of extracellular matrix proteins in osteoarthritic (OA) articular cartilage. Thus, catabolism results in the elevated loss of extracellular matrix proteins. There is also evidence of an increase in the frequency of chondrocyte apoptosis that compromises the capacity of articular cartilage to undergo repair. Although much of the fundamental OA studies over the past 20 years identified and characterized many genes relevant to pro-inflammatory cytokines, apoptosis, and matrix metalloproteinases (MMPs)/a disintegrin and metalloproteinase with thrombospondin motif (ADAMTS), more recent studies focused on epigenetic mechanisms and the associated role of microRNAs (miRs) in regulating gene expression in OA cartilage. Thus, several miRs were identified as regulators of chondrocyte signaling pathways, apoptosis, and proteinase gene expression. For example, the reduced expression of miR-146a was found to be coupled to reduced type II collagen (COL2) in OA cartilage, whereas MMP-13 levels were increased, suggesting an association between MMP-13 gene expression and COL2A1 gene expression. Results of these studies imply that microRNAs could become useful in the search for diagnostic biomarkers, as well as providing novel therapeutic targets for intervention in OA.
Collapse
Affiliation(s)
- Charles J Malemud
- Department of Medicine, Division of Rheumatic Diseases, University Hospitals Cleveland Medical Center, Foley Medical Building, 2061 Cornell Road, Cleveland, OH 44106-5076, USA.
| |
Collapse
|
40
|
Sun H, Zhao X, Zhang C, Zhang Z, Lun J, Liao W, Zhang Z. MiR-455-3p inhibits the degenerate process of chondrogenic differentiation through modification of DNA methylation. Cell Death Dis 2018; 9:537. [PMID: 29748607 PMCID: PMC5945650 DOI: 10.1038/s41419-018-0565-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/19/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
The aim of this work was to determine whether miR-455-3p regulates DNA methylation during chondrogenic differentiation of hMSCs. The expression of miR-455-3p and de novo methyltransferase DNMT3A was assessed in micromass culture of hBMSCs, which induced chondrogenic differentiation in vitro, and in E16.5 mice in vivo. A luciferase reporter assay was used to confirm whether miR-455-3p directly targets DNMT3A by interaction with the 3′-UTR. Using an Illumina Infinium Methylation EPIC microarray, genome-wide DNA methylation of hBMSCs with or without overexpressed miR-455-3p was examined for 28 days during induced chondrogenic differentiation. Here, we showed that miR-455-3p was more expressed during the middle stage of hBMSC chondrogenic differentiation, and less expressed in the late stage. DNMT3A was less expressed in the middle stage and more expressed in the late stage, and was also more expressed in the palms of miR-455-3p deletion mice compared to those of wild-type mice. The luciferase reporter assay demonstrated that miR-455-3p directly targets DNMT3A 3′-UTR. miR-455-3p overexpression inhibits the degenerate process during chondrogenic differentiation, while deletion of miR-455-3p in mice accelerated cartilage degeneration. Genome-wide DNA methylation analysis showed miR-455-3p overexpression regulates DNA methylation of cartilage-specific genes. GO analysis revealed PI3K-Akt signaling pathway was most hypomethylated. Our data show that miR-455-3p can regulate hMSC chondrogenic differentiation by affecting DNA methylation. Overexpression of miR-455-3p and DNA methylation inhibitors can thus potentially be utilized to optimize chondrogenic differentiation.
Collapse
Affiliation(s)
- Hao Sun
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Xiaoyi Zhao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Chengyun Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Ziji Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Jiayong Lun
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China
| | - Weiming Liao
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China.
| | - Zhiqi Zhang
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, 510080, Guangzhou, Guangdong, China.
| |
Collapse
|
41
|
Chen BB, Li ZH, Gao S. Circulating miR-146a/b correlates with inflammatory cytokines in COPD and could predict the risk of acute exacerbation COPD. Medicine (Baltimore) 2018; 97:e9820. [PMID: 29443743 PMCID: PMC5839872 DOI: 10.1097/md.0000000000009820] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to investigate the predicting value of miR-146a/b for acute exacerbation chronic obstructive pulmonary disease (AECOPD) and COPD, and to explore their associations with inflammatory cytokines in AECOPD and stable COPD patients.One hundred six AECOPD, 122 stable COPD patients, and 110 health volunteers with age and sex matched to total COPD patients (AECOPD and stable COPD) were enrolled. Blood samples were collected from all participants. Relative expression of miR-146a/b was determined by real-time polymerase chain reaction. Tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), leukotriene B4 (LTB-4) expression in serum from AECOPD and stable COPD patients were assessed using commercial ELISA kit.Serum levels of miR-146a and miR-146b were down regulated in AECOPD patients compared with stable COPD patients and HCs. miR-146a and miR-146b are of good values for predicting the risk of AECOPD in HCs with AUC of 0.702 and 0.715. Additionally, miR-146a and miR-146b could distinguish AECOPD from stable COPD patients with AUC of 0.670 and 0.643. In AECOPD patients, levels of miR-146a in AECOPD patients were negatively associated with TNF-α, IL-6, IL-8, and LTE-4 expression. In stable COPD patients, miR-146a expressions were negatively correlated with TNF-α, IL-1β, IL-6, IL-8, and LTE-4 levels. And, the expressions of miR-146b in AECOPD patients were negatively associated with IL-1β and LTB-4 expression. While in stable COPD patients, miR-146b expressions were only negatively correlated with TNF-α level.In conclusion, miR-146a and miR-146b were negatively correlated with inflammatory cytokines, and could be promising biomarkers for predicting the risk of AECOPD in stable COPD patients and healthy individuals.
Collapse
|
42
|
Abstract
INTRODUCTION Osteoarthritis (OA), a chronic, debilitating and degenerative disease of the joints, is the most common form of arthritis. The seriousness of this prevalent and chronic disease is often overlooked. Disease modifying OA drug development is hindered by the lack of soluble biomarkers to detect OA early. The objective of OA biomarker research is to identify early OA prior to the appearance of radiographic signs and the development of pain. Areas covered: This review has focused on extracellular genomic material that could serve as biomarkers of OA. Recent studies have examined the expression of extracellular genomic material such as miRNA, lncRNA, snoRNA, mRNA and cell-free DNA, which are aberrantly expressed in the body fluids of OA patients. Changes in genomic content of peripheral blood mononuclear cells in OA could also function as biomarkers of OA. Expert commentary: There is an unmet need for soluble biomarkers for detecting and then monitoring OA disease progression. Extracellular genomic material research may also reveal more about the underlying pathophysiology of OA. Minimally-invasive liquid biopsies such as synovial fluid and blood sampling of genomic material may be more sensitive over radiography in the detection, diagnosis and monitoring of OA in the future.
Collapse
Affiliation(s)
- Emma Budd
- a The D-BOARD European Consortium for Biomarker Discovery, School of Veterinary Medicine , University of Surrey , Guildford , UK.,b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| | - Giovanna Nalesso
- b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK
| | - Ali Mobasheri
- a The D-BOARD European Consortium for Biomarker Discovery, School of Veterinary Medicine , University of Surrey , Guildford , UK.,b Department of Veterinary Pre-Clinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences , University of Surrey , Guildford , UK.,c Arthritis Research UK Centre for Sport, Exercise and Osteoarthritis , Queen's Medical Centre , Nottingham , UK
| |
Collapse
|
43
|
Budd E, Waddell S, de Andrés MC, Oreffo ROC. The Potential of microRNAs for Stem Cell-based Therapy for Degenerative Skeletal Diseases. ACTA ACUST UNITED AC 2017; 3:263-275. [PMID: 29214143 PMCID: PMC5700219 DOI: 10.1007/s40610-017-0076-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of Review Degenerative skeletal disorders including osteoarthritis (OA) and osteoporosis (OP) are the result of attenuation of tissue regeneration and lead to painful conditions with limited treatment options. Preventative measures to limit the onset of OA and OP remain a significant unmet clinical need. MicroRNAs (miRNAs) are known to be involved in the differentiation of stem cells, and in combination with stem cell therapy could induce skeletal regeneration and potentially prevent OA and OP onset. Recent Findings The combination of stem cells and miRNA has been successful at regenerating the bone and cartilage in vivo. MiRNAs, including miR-146b known to be involved in chondrogenic differentiation, could provide innovative targets for stem cell-based therapy, for the repair of articular cartilage defects forestalling the onset of OA or in the generation of a stem cell-based therapy for OP. Summary This review discusses the combination of skeletal stem cells (SSCs) and candidate miRNAs for application in a cell-based therapy approach for skeletal regenerative medicine.
Collapse
Affiliation(s)
- Emma Budd
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Shona Waddell
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - María C de Andrés
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Richard O C Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|