1
|
Hung CY, Hsueh TY, Rethi L, Lu HT, Chuang AEY. Advancements in regenerative medicine: a comprehensive review of stem cell and growth factor therapies for osteoarthritis. J Mater Chem B 2025; 13:4494-4526. [PMID: 40042377 DOI: 10.1039/d4tb01769b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Osteoarthritis (OA) is a widely encountered degenerative joint disorder marked by gradual cartilage deterioration, inflammation, and pain, which collectively impose considerable strain on global healthcare systems. While traditional therapies typically offer relief from symptoms, they do not tackle the core pathophysiological aspects of the disease. Regenerative medicine has recently risen as a promising field for addressing OA, capitalizing on the regenerative capabilities of stem cells and growth factors to foster tissue healing and renewal. This thorough review delves into the most recent progress in stem cell and growth factor treatments for OA, covering preclinical studies, clinical trials, and novel technological developments. We discuss the diverse origins of stem cells, such as mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and adipose-derived stem cells (ASCs), underscoring their therapeutic actions and effectiveness in both preclinical and clinical environments. Moreover, we explore contributions of growth factors like transforming growth factor (TGF)-β, platelet-derived growth factor (PDGF), and insulin-like growth factor (IGF) in modifying OA's pathology and enhancing tissue restoration. Additionally, this review discusses the hurdles and constraints tied to current regenerative strategies, including the standardization of cell sources, the refinement of delivery techniques, and considerations for long-term safety. By meticulously assessing the latest research outcomes and technological breakthroughs, this review aims to shed light on the potential of stem cell and growth factor therapies as forthcoming therapeutic options for OA, thereby propelling forward the domain of regenerative medicine and enhancing clinical results for individuals afflicted with this incapacitating ailment.
Collapse
Affiliation(s)
- Chen-Yuan Hung
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tai-Yuan Hsueh
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lekshmi Rethi
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
| | - Hsien-Tsung Lu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei City 11031, Taiwan
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- International PhD Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Andrew E-Y Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
- Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Padhee S, Mohanty D, Sahoo A, Jena S, Chandra Panda P, Ray A, Nayak S. Exploring the mechanism of action of Vanda tessellata extract for the treatment of osteoarthritis through network pharmacology, molecular modelling and experimental assays. Heliyon 2024; 10:e35971. [PMID: 39224251 PMCID: PMC11367146 DOI: 10.1016/j.heliyon.2024.e35971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The present study employed a comprehensive approach of network pharmacology, molecular dynamic simulation and in-vitro assays to investigate the underlying mechanism of the anti-osteoarthritic potential of Vanda tessellata extract (VTE). Thirteen active compounds of VTE were retrieved from the literature and the IMPPAT database. All of these passed the drug likeness and oral bioavailability parameters. A total of 535 VTE targets and 2577 osteoarthritis related targets were obtained. The compound-target-disease network analysis revealed vanillin, daucosterol, gigantol and syringaldehyde as the core key components. Protein-protein interaction analysis revealed BCL2, FGF2, ICAM 1, MAPK1, MMP1, MMP2, MMP9, COX2, STAT3 and ESR1 as the hub genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed AGE-RAGE signalling pathway, HIF-1 signalling pathway and ESR signalling pathway as the major signalling pathway of VTE involved in treating osteoarthritis. Molecular docking analysis showed daucosterol and gigantol to have good binding affinity with BCL2, ESR1 and MMP9, and the results were further confirmed through molecular dynamics simulation analysis. The mechanism predicted by network pharmacology was validated in vitro on IL-1β-induced SW982 synovial cells. VTE did not show any cytotoxicity and inhibited the migration of SW982 cells. VTE inhibited the expression level of IL-6, IL-8, TNF-α, PGE-2, MMP-2 and MMP-9 in a dose-dependent manner. VTE inhibited nuclear translocation of NF- κβ and suppressed phosphorylation of p38, extracellular signal-regulated kinase (ERK), and c-Jun NH2-terminal kinase (JNK) of the mitogen-activated protein kinase (MAPK) signalling pathway. The results showed that VTE exerted an anti-osteoarthritic effect by a multi-target, multi-component and multi-signalling pathway approach.
Collapse
Affiliation(s)
- Sucheesmita Padhee
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| | - Debajani Mohanty
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| | - Ambika Sahoo
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| | - Sudipta Jena
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| | - Pratap Chandra Panda
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| | - Asit Ray
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| | - Sanghamitra Nayak
- Centre for Biotechnology, Siksha O Anusandhan (Deemed to be University), Kalinganagar, Ghatikia, Bhubaneswar, India
| |
Collapse
|
3
|
Wolfe PN, Stoker AM, Leary E, Crist BD, Bozynski CC, Cook JL. Evaluation of Serum and Urine Biomarker Panels for Developmental Dysplasia of the Hip Prior to Onset of Secondary Osteoarthritis. Cartilage 2024; 15:164-174. [PMID: 37051936 PMCID: PMC11368892 DOI: 10.1177/19476035231163032] [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: 01/13/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 04/14/2023] Open
Abstract
OBJECTIVE Evaluate serum and urine biomarker panels for their capabilities in discriminating between individuals (13- to 34-years-olds) with healthy hips versus those with developmental dysplasia of the hip (DDH) prior to diagnosis of secondary hip osteoarthritis (OA). DESIGN Urine and serum were collected from individuals (15-33 years old) with DDH, prior to and following diagnosis of hip OA, and from age-matched healthy-hip controls. Samples were analyzed for panels of protein biomarkers with potential for differentiation of hip status using receiver operator characteristic curve (area under curve [AUC]) assessments. RESULTS Multiple urine and serum biomarker panels effectively differentiated individuals with DDH from healthy-hip controls in a population at risk for developing secondary hip OA with the best performing panel demonstrating an AUC of 0.959. The panel comprised of two serum and two urinary biomarkers provided the highest combined values for sensitivity, 0.85, and specificity, 1.00, while a panel of four serum biomarkers provided the highest sensitivity, 0.93, while maintaining adequate specificity, 0.71. CONCLUSION Results of this study indicate that panels of protein biomarkers measured in urine and serum may be able to differentiate young adults with DDH from young adults with healthy hips. These data suggest the potential for clinical application of a routine diagnostic method for cost-effective and timely screening for DDH in at-risk populations. Further development and validation of these biomarker panels may result in highly sensitive and specific tools for early diagnosis, staging, and prognostication of DDH, as well as treatment decision making and monitoring capabilities. LEVEL OF EVIDENCE III.
Collapse
Affiliation(s)
- Preston N. Wolfe
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - Aaron M. Stoker
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - Emily Leary
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
- School of Medicine, University of Missouri, Columbia, MO, USA
| | - Brett D. Crist
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| | - Chantelle C. Bozynski
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
| | - James L. Cook
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, USA
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, USA
| |
Collapse
|
4
|
Jia S, Liang R, Chen J, Liao S, Lin J, Li W. Emerging technology has a brilliant future: the CRISPR-Cas system for senescence, inflammation, and cartilage repair in osteoarthritis. Cell Mol Biol Lett 2024; 29:64. [PMID: 38698311 PMCID: PMC11067114 DOI: 10.1186/s11658-024-00581-x] [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: 12/29/2023] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Osteoarthritis (OA), known as one of the most common types of aseptic inflammation of the musculoskeletal system, is characterized by chronic pain and whole-joint lesions. With cellular and molecular changes including senescence, inflammatory alterations, and subsequent cartilage defects, OA eventually leads to a series of adverse outcomes such as pain and disability. CRISPR-Cas-related technology has been proposed and explored as a gene therapy, offering potential gene-editing tools that are in the spotlight. Considering the genetic and multigene regulatory mechanisms of OA, we systematically review current studies on CRISPR-Cas technology for improving OA in terms of senescence, inflammation, and cartilage damage and summarize various strategies for delivering CRISPR products, hoping to provide a new perspective for the treatment of OA by taking advantage of CRISPR technology.
Collapse
Affiliation(s)
- Shicheng Jia
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Rongji Liang
- Shantou University Medical College, Shantou, 515041, China
| | - Jiayou Chen
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shantou University Medical College, Shantou, 515041, China
| | - Shuai Liao
- Department of Bone and Joint, Peking University Shenzhen Hospital, Shenzhen, 518036, China
- Shenzhen University School of Medicine, Shenzhen, 518060, China
| | - Jianjing Lin
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| | - Wei Li
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
5
|
Juma SN, Liao J, Huang Y, Vlashi R, Wang Q, Wu B, Wang D, Wu M, Chen G. Osteoarthritis versus psoriasis arthritis: Physiopathology, cellular signaling, and therapeutic strategies. Genes Dis 2024; 11:100986. [PMID: 38292181 PMCID: PMC10825447 DOI: 10.1016/j.gendis.2023.04.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/15/2023] [Indexed: 02/01/2024] Open
Abstract
Osteoarthritis and psoriasis arthritis are two degenerative forms of arthritis that share similar yet also different manifestations at the histological, cellular, and clinical levels. Rheumatologists have marked them as two entirely distinct arthropathies. Given recent discoveries in disease initiation and progression, potential mechanisms, cellular signaling pathways, and ongoing clinical therapeutics, there are now more opportunities for discovering osteoarthritis drugs. This review summarized the osteoarthritis and psoriasis arthritis signaling pathways, crosstalk between BMP, WNT, TGF-β, VEGF, TLR, and FGF signaling pathways, biomarkers, and anatomical pathologies. Through bench research, we demonstrated that regenerative medicine is a promising alternative for treating osteoarthritis by highlighting significant scientific discoveries on entheses, multiple signaling blockers, and novel molecules such as immunoglobulin new antigen receptors targeted for potential drug evaluation. Furthermore, we offered valuable therapeutic approaches with a multidisciplinary strategy to treat patients with osteoarthritis or psoriasis arthritis in the coming future in the clinic.
Collapse
Affiliation(s)
- Salma Nassor Juma
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Qingwan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Bocong Wu
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Dan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| | - Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, China
| |
Collapse
|
6
|
Zhao Y, Gan YH. Combination of hyperlipidemia and 17β-Estradiol induces TMJOA-like pathological changes in rats. Oral Dis 2023; 29:3640-3653. [PMID: 35765240 DOI: 10.1111/odi.14294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/12/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE We explored whether hyperlipidemia or combination of hyperlipidemia and E2 could induce TMJOA. MATERIALS AND METHODS Four groups of female rats were treated with normal diet, normal diet with E2, high-fat diet, and high-fat diet with E2 (HFD/E2), respectively, to induce TMJOA till 8 weeks. Another three groups were then used for COX2 inhibitor celecoxib to block the induction of TMJOA. Primary condylar chondrocytes were treated with combination of E2, ox-LDL, and corresponding inhibitors for evaluating expressions of related molecules. RESULTS Condylar cartilage proliferation with plenty of chondrocyte apoptosis and increased staining for LOX1, nuclear NF-κB, IL-1β, and COX2 at 4 weeks and decreased condylar cartilage and increased subchondral bone density at 8 weeks were observed only in the HFD/E2 group. Celecoxib significantly alleviated the cartilage proliferation and apoptosis in the HFD/E2 group. Serum ox-LDL increased in both high-fat diet groups, while serum IL-1β increased only in the HFD/E2 group. Combination of E2 and ox-LDL synergistically induced expressions of LOX1, phosphorylated NF-κB, IL-1β, and COX2, while LOX1 inhibitor blocked the induction of phosphorylated NF-κB, and NF-κB inhibitor the induction of IL-1β, and IL-1β inhibitor the induction of COX2. CONCLUSION Combination of hyperlipidemia and E2-induced TMJOA-like pathological changes through LOX1/NF-κB/IL-1β/COX2-signaling pathway.
Collapse
Affiliation(s)
- Yan Zhao
- Central laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Oral & Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ye-Hua Gan
- Central laboratory, Peking University School and Hospital of Stomatology, Beijing, China
- Department of Oral & Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
- Center for Temporomandibular Disorders & Orofacial Pain, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
7
|
Cavallo C, Boffa A, Salerno M, Merli G, Grigolo B, Filardo G. Adipose Tissue-Derived Products May Present Inflammatory Properties That Affect Chondrocytes and Synoviocytes from Patients with Knee Osteoarthritis. Int J Mol Sci 2023; 24:12401. [PMID: 37569775 PMCID: PMC10418602 DOI: 10.3390/ijms241512401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Adipose tissue-derived cell-based injectable therapies have been demonstrated to have disease-modifying effects on joint tissues in preclinical studies on animal osteoarthritis (OA) models, but clinical results are heterogeneous and not always satisfactory. The aim of this study was to investigate the influence of adipose tissue properties on the therapeutic effects of the adipose-derived product in an in vitro OA setting. Micro-fragmented adipose tissue (MF-AT) samples were obtained from 21 OA patients (mean age 51.7 ± 11.8 years, mean BMI 25.7 ± 4.1 kg/m2). The analysis of the MF-AT supernatant was performed to analyze the release of inflammatory factors. The effects of MF-AT inflammatory factors were investigated on chondrocytes and synoviocytes gene expression levels. Patients' characteristics were analyzed to explore their influence on MF-AT inflammatory molecules and on the MF-AT effects on the gene expression of chondrocytes and synoviocytes. The study results demonstrated that adipose tissue-derived products may present inflammatory properties that influence the therapeutic potential for OA treatment, with products with a higher pro-inflammatory profile stimulating a higher expression of genes related to a more inflamed and catabolic phenotype. A higher pro-inflammatory cytokine pattern and a higher pro-inflammatory effect were found in adipose tissue-derived products obtained from OA patients with higher BMI.
Collapse
Affiliation(s)
- Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (C.C.); (B.G.)
| | - Angelo Boffa
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Manuela Salerno
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.M.); (G.F.)
| | - Giulia Merli
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.M.); (G.F.)
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (C.C.); (B.G.)
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.M.); (G.F.)
| |
Collapse
|
8
|
Wolfe PN, Stoker AM, Crist BD, Leary E, Bozynski CC, Cook JL. Evaluation of mechanistic serum and urine biomarkers for secondary osteoarthritis associated with developmental dysplasia of the hip. J Orthop 2023; 42:24-29. [PMID: 37601815 PMCID: PMC10435905 DOI: 10.1016/j.jor.2023.05.010] [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: 03/16/2023] [Revised: 05/09/2023] [Accepted: 05/24/2023] [Indexed: 08/22/2023] Open
Abstract
Objective Determine measurable differences for mechanistic urine and serum biomarkers in patients with developmental dysplasia of the hip (DDH) prior to, and following, secondary hip osteoarthritis (OA) when compared to controls. Design Urine and serum were collected from individuals with developmental dysplasia of the hip (n = 39), prior to (Pre-OA DDH, n = 32) and following diagnosis of secondary hip OA (Post-OA DDH, n = 7), age-matched Pre-OA controls (n = 35), and age-matched Post-OA controls (n = 12). Samples were analyzed for protein biomarkers with potential for differentiation of hip status through a Mann-Whitney U test with a Benjamini-Hochberg correction. Results Several interleukin and degradation related proteins were found to be differentially expressed when comparing DDH-related hip status prior to and following diagnosis of hip OA. In addition, MCP-1 and TIMP-1 were significantly different between younger and older patients in the control cohorts. Conclusion These results provide initial evidence for serum and urine protein biomarkers that define clinically relevant stages of symptomatic DDH and its progression to secondary hip osteoarthritis categorized by known mechanisms of disease. Level of evidence III.
Collapse
Affiliation(s)
- Preston N. Wolfe
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
- Arthrex, Inc, Naples, FL, United States
| | - Aaron M. Stoker
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - Brett D. Crist
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - Emily Leary
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - Chantelle C. Bozynski
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| | - James L. Cook
- Department of Orthopaedic Surgery, University of Missouri, Columbia, MO, United States
- Thompson Laboratory for Regenerative Orthopaedics, University of Missouri, Columbia, MO, United States
| |
Collapse
|
9
|
Michelacci YM, Baccarin RYA, Rodrigues NNP. Chondrocyte Homeostasis and Differentiation: Transcriptional Control and Signaling in Healthy and Osteoarthritic Conditions. Life (Basel) 2023; 13:1460. [PMID: 37511835 PMCID: PMC10381434 DOI: 10.3390/life13071460] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/13/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Chondrocytes are the main cell type in articular cartilage. They are embedded in an avascular, abundant, and specialized extracellular matrix (ECM). Chondrocytes are responsible for the synthesis and turnover of the ECM, in which the major macromolecular components are collagen, proteoglycans, and non-collagen proteins. The crosstalk between chondrocytes and the ECM plays several relevant roles in the regulation of cell phenotype. Chondrocytes live in an avascular environment in healthy cartilage with a low oxygen supply. Although chondrocytes are adapted to anaerobic conditions, many of their metabolic functions are oxygen-dependent, and most cartilage oxygen is supplied by the synovial fluid. This review focuses on the transcription control and signaling responsible for chondrocyte differentiation, homeostasis, senescence, and cell death and the changes that occur in osteoarthritis. The effects of chondroitin sulfate and other molecules as anti-inflammatory agents are also approached and analyzed.
Collapse
Affiliation(s)
- Yara M Michelacci
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, SP, Brazil
| | - Raquel Y A Baccarin
- Faculdade de Medicina Veterinária e Zootecnia, Universidade São Paulo, São Paulo 05508-270, SP, Brazil
| | - Nubia N P Rodrigues
- Faculdade de Medicina Veterinária e Zootecnia, Universidade São Paulo, São Paulo 05508-270, SP, Brazil
| |
Collapse
|
10
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
11
|
Yang L, Liu J, Yu Y, Liu S. A novel signature of autophagy-related immunophenotyping biomarkers in osteoarthritis. Life Sci 2023; 321:121599. [PMID: 36966915 DOI: 10.1016/j.lfs.2023.121599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 04/03/2023]
Abstract
AIMS We aimed to provide an autophagy-related signature to seek immunophenotyping biomarkers in osteoarthritis (OA). MATERIALS AND METHODS Microarray expression profiling of OA subchondral bone samples and screening of an autophagy database for autophagy-related differentially expressed genes (au-DEGs) between OA and normal samples were performed. A weighted gene co-expression network analysis (WGCNA) was constructed using au-DEGs to identify key modules significantly associated with clinical information of OA samples. OA-related autophagy hub genes were identified based on the connectivity with the phenotypes of genes in key modules and the protein-protein interaction (PPI) network in which the genes in the modules are involved, followed by feasibility verification of autophagy hub genes by bioinformatics analysis and biological experiments. KEY FINDINGS We screened 754 au-DEGs between OA and control samples, and co-expression networks were constructed using au-DEGs. Three OA-related autophagy hub genes (HSPA5, HSP90AA1, and ITPKB) were identified. Based on the hub gene expression profiles, OA samples were divided into two clusters with significantly different expression profiles and distinct immunological features, and the three hub genes were significantly differentially expressed between the clusters. Differences in hub genes between OA and control samples regarding sex, age, and grades of OA were examined using external datasets and experimental validation. SIGNIFICANCE Three autophagy-related markers of OA were identified using bioinformatics methods, and these markers may be useful for the autophagy-related immunophenotyping of OA. The present data may facilitate the diagnosis of OA, as well as the design of immunotherapies and individualized medical treatments.
Collapse
Affiliation(s)
- Liyu Yang
- Department of Orthopedics, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China.
| | - Jiamei Liu
- Department of Pathology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China.
| | - Yuanqi Yu
- Department of Clinical Medicine, Innovation Institute of China Medical University, Shenyang, Liaoning 110122, PR China.
| | - Shengye Liu
- Department of Orthopedics, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, PR China.
| |
Collapse
|
12
|
Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S, Xiao G. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther 2023; 8:56. [PMID: 36737426 PMCID: PMC9898571 DOI: 10.1038/s41392-023-01330-w] [Citation(s) in RCA: 449] [Impact Index Per Article: 224.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder that leads to disability and affects more than 500 million population worldwide. OA was believed to be caused by the wearing and tearing of articular cartilage, but it is now more commonly referred to as a chronic whole-joint disorder that is initiated with biochemical and cellular alterations in the synovial joint tissues, which leads to the histological and structural changes of the joint and ends up with the whole tissue dysfunction. Currently, there is no cure for OA, partly due to a lack of comprehensive understanding of the pathological mechanism of the initiation and progression of the disease. Therefore, a better understanding of pathological signaling pathways and key molecules involved in OA pathogenesis is crucial for therapeutic target design and drug development. In this review, we first summarize the epidemiology of OA, including its prevalence, incidence and burdens, and OA risk factors. We then focus on the roles and regulation of the pathological signaling pathways, such as Wnt/β-catenin, NF-κB, focal adhesion, HIFs, TGFβ/ΒΜP and FGF signaling pathways, and key regulators AMPK, mTOR, and RUNX2 in the onset and development of OA. In addition, the roles of factors associated with OA, including MMPs, ADAMTS/ADAMs, and PRG4, are discussed in detail. Finally, we provide updates on the current clinical therapies and clinical trials of biological treatments and drugs for OA. Research advances in basic knowledge of articular cartilage biology and OA pathogenesis will have a significant impact and translational value in developing OA therapeutic strategies.
Collapse
Affiliation(s)
- Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
13
|
Two Modulators of Skeletal Development: BMPs and Proteoglycans. J Dev Biol 2022; 10:jdb10020015. [PMID: 35466193 PMCID: PMC9036252 DOI: 10.3390/jdb10020015] [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: 02/01/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022] Open
Abstract
During embryogenesis, skeletal development is tightly regulated by locally secreted growth factors that interact with proteoglycans (PGs) in the extracellular matrix (ECM). Bone morphogenetic proteins (BMPs) are multifunctional growth factors that play critical roles in cartilage maturation and bone formation. BMP signals are transduced from plasma membrane receptors to the nucleus through both canonical Smad and noncanonical p38 mitogen-activated protein kinase (MAPK) pathways. BMP signalling is modulated by a variety of endogenous and exogenous molecular mechanisms at different spatiotemporal levels and in both positive and negative manners. As an endogenous example, BMPs undergo extracellular regulation by PGs, which generally regulate the efficiency of ligand-receptor binding. BMP signalling can also be exogenously perturbed by a group of small molecule antagonists, such as dorsomorphin and its derivatives, that selectively bind to and inhibit the intracellular kinase domain of BMP type I receptors. In this review, we present a current understanding of BMPs and PGs functions in cartilage maturation and osteoblast differentiation, highlighting BMP–PG interactions. We also discuss the identification of highly selective small-molecule BMP receptor type I inhibitors. This review aims to shed light on the importance of BMP signalling and PGs in cartilage maturation and bone formation.
Collapse
|
14
|
Liu C, Chen Y. Ketorolac tromethamine alleviates IL‑1β‑induced chondrocyte injury by inhibiting COX‑2 expression. Exp Ther Med 2022; 23:337. [PMID: 35411209 PMCID: PMC8992376 DOI: 10.3892/etm.2022.11267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/19/2021] [Indexed: 11/27/2022] Open
Abstract
Osteoarthritis (OA) is one of the most frequently diagnosed chronic diseases, and its prevalence is rising as life expectancy increases. The present study was designed to investigate the role of ketorolac tromethamine (KT) in OA by establishing an in vitro model in ATDC5 cells. The OA model was established through induction using 10 ng/ml IL-1β. KT was then used to treat the ATDC5 cells. An MTT assay was adopted to detect the viability of ATDC5 cells with or without IL-1β induction, and cyclo-oxygenase-2 (COX-2) expression in IL-1β-induced ATDC5 cells was measured via reverse transcription-quantitative (RT-q)PCR and western blotting. To explore the effects of KT on proliferation and apoptosis in IL-1β-induced ATDC5 cells, COX-2 was overexpressed and RT-qPCR was employed to detect the mRNA expression of COX-2. The viability of IL-1β-induced ATDC5 cells was detected by using a Cell Counting Kit-8 assay. In addition, levels of apoptosis and apoptosis-related proteins were determined using TUNEL staining and western blotting, respectively. Additionally, the effects of KT on oxidative stress in IL-1β-induced ATDC5 cells were also investigated. The expression levels of nitric oxide (NO) and inducible NO synthase (iNOS) were detected via NO kit assay and western blotting, respectively. In addition, the expression levels of oxidative stress-related proteins, including reactive oxygen species (ROS), superoxide dismutase (SOD) and prostaglandin E2 (PGE2), were determined using ELISA. To investigate the effects of KT on the inflammatory response and extracellular matrix (ECM) degradation, ELISA and western blotting were adopted to detect inflammatory-related proteins and ECM degradation-related proteins. Results from MTT assay indicated that KT decreased ATDC5 cell viability in a concentration-dependent manner. The expression of COX-2 was found to be downregulated in IL-1β-induced ATDC5 cells after treatment with KT, according to RT-qPCR and western blotting results. KT inhibited apoptosis and the expression levels of NO, iNOS and inflammatory-related proteins in IL-1β-induced ATDC5 cells, while COX-2 overexpression reversed these inhibitory effects. However, the increased proliferation of IL-1β-induced ATDC5 cells after the stimulation of KT was decreased by COX-2 overexpression. Additionally, KT upregulated Bcl-2, SOD, type II collagen and aggrecan expression levels in IL-1β-induced ATDC5 cells, whereas Bax, ROS, matrix metallopeptidase (MMP)1 and MMP13 expression levels were downregulated. KT promoted the proliferation of IL-1β-induced ATDC5 cells, whereas COX-2 overexpression reversed the promotive effects of KT, revealing that KT could alleviate IL-1β-induced chondrocyte injury by suppressing COX-2 expression.
Collapse
Affiliation(s)
- Chang Liu
- Orthopedic Diagnosis and Center of Huating First People's Hospital, Pingliang, Gansu 744100, P.R. China
| | - Yizhi Chen
- Department of Orthopedics and Traumatology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian 364000, P.R. China
| |
Collapse
|
15
|
Zhao Z, Wang Z, Pei L, Zhou X, Liu Y. Long non-coding ribonucleic acid AFAP1-AS1 promotes chondrocyte proliferation via the miR-512-3p/matrix metallopeptidase 13 (MMP-13) axis. Bioengineered 2022; 13:5386-5395. [PMID: 35188875 PMCID: PMC8973689 DOI: 10.1080/21655979.2022.2031390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Long-chain non-coding RNAs are reported to be involved in cartilage damage. However, less research on the role of actin filament-associated protein 1 antisense RNA 1 (AFAP1-AS1) in osteoarthritis. To investigate AFAP1-AS1 function in osteoarthritis development, AFAP1-AS1 and miR-512-3p expression levels in osteoarthritis cartilage and cells were evaluated using RT-qPCR. The downstream target genes of AFAP1-AS1 and miR-512-3p were predicted and validated using luciferase reporter assays. Moreover, a knee osteoarthritis model was established by injecting monoiodoacetate into the knee joints of mice. The effects of AFAP1-AS1 and miR-512-3p on osteoarthritis chondrocyte proliferation and MMP-13, collagen II, and collagen IV expressions were detected in vivo using CCK-8 assay and Western blotting and RT-qPCR, respectively. AFAP1-AS1 expression was upregulated in osteoarthritis cartilage and cells. MiR-512-3p expression was downregulated in osteoarthritis cartilage. AFAP1-AS1 overexpression inhibited miR-512-3p expression in chondrocytes. Furthermore, AFAP1-AS1 over-expression promoted chondrocyte proliferation, and miR-512-3p mimic inhibited chondrocyte proliferation in vivo. AFAP1-AS1 overexpression reduced type II and type IV collagen expression, while miR-512-3p overexpression promoted type II and type IV collagen in vivo. AFAP1-AS1 overexpression enhanced MMP-13 expression in vivo. AFAP1-AS1 overexpression regulated chondrocyte proliferation by inhibiting miR-512-3p expression in vivo. AFAP1-AS1 could be a potential target to treat osteoarthritis by inhibiting miR-512-3p and subsequently inducing chondrocyte proliferation and regulating matrix synthesis.
Collapse
Affiliation(s)
- Zhi Zhao
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, P. R. China
| | - Zhiyan Wang
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, P. R. China
| | - Lijia Pei
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, P. R. China
| | - Xinshe Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, P. R. China
| | - Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, P. R. China
| |
Collapse
|
16
|
Liu Y, Liu J, Ma Y, Zhang Y, Chen Q, Yang X, Shang Y. The protective effects of Olmesartan against interleukin-29 (IL-29)-induced type 2 collagen degradation in human chondrocytes. Bioengineered 2022; 13:1802-1813. [PMID: 35012432 PMCID: PMC8805962 DOI: 10.1080/21655979.2021.1997090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/19/2021] [Indexed: 12/31/2022] Open
Abstract
Osteoarthritis (OA) is a cartilage degenerative disease commonly observed in the elderly population and is pathologically characterized by the degradation of the cartilage extracellular matrix (ECM). Matrix metalloproteinases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs) are critical enzymes involved in the degradation of ECM. Olmesartan is an inhibitor of the angiotensin II receptor developed for the treatment of hypertension, and recent studies show that it exerts anti-inflammatory effects in arthritis. The present study aimed to investigate the mechanism of the protective effect of Olmesartan on cartilage ECM degradation. Interleukin-29 (IL-29) is a novel inflammatory mediator involved in the inflammation and degradation of cartilage in OA, and human T/C-28a2 cells treated with it were the inflammatory model in vitro. We found that the degradation of type 2 collagens and aggrecans was induced by IL-29, accompanied by the upregulation of MMPs and ADAMTSs, but the presence of Olmesartan significantly ameliorated these increases. In addition, Olmesartan abolished IL-29- induced oxidative stress and elevated the expression level of TNF receptor-associated factor 6 (TRAF-6). Mechanistically, we showed that Olmesartan suppressed IL-29- caused inhibitor kappa B α (IκBα) expression and nuclear translocation of nuclear factor kappa-B (NF-κB) p65, indicating it suppressed the activation of the NF-κB pathway. Collectively, our data reveal that Olmesartan exerted a protective function on IL-29- induced type 2 collagen degradation in human chondrocytes.
Collapse
Affiliation(s)
- Yunlong Liu
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Junyi Liu
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Yan Ma
- Lab of Molecular Biology, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Yongyong Zhang
- Lab of Molecular Biology, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Qiong Chen
- Department of Medicine, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Xin Yang
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| | - Yanchun Shang
- Department of Knee Surgery, Luoyang Orthopedic-Traumatological Hospital of Henan Province, Zhengzhou, China
| |
Collapse
|
17
|
wei W, He S, Wang Z, Dong J, Xiang D, Li Y, Ren L, Kou N, Lv J. LINC01534 Promotes the Aberrant Metabolic Dysfunction and Inflammation in IL-1β-Simulated Osteoarthritic Chondrocytes by Targeting miR-140-5p. Cartilage 2021; 13:898S-907S. [PMID: 31735077 PMCID: PMC8804787 DOI: 10.1177/1947603519888787] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Long non-coding RNA 01534 (LINC01534) is highly expressed in the tissues of patients with osteoarthritis (OA). This study investigated the mechanism of LINC01534 on abnormal metabolic dysfunction in OA chondrocytes induced by interleukin-1β (IL-1β). METHODS The quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the expressions of LINC01534, aggrecan, collagen II, and matrix metalloproteinase (MMPs) in OA cartilage tissue or OA chondrocyte model induced by IL-1β. The expressions of aggrecan and collagen II in the chondrocyte were detected by Western blot. The levels of tumor necrosis factor-α (TNF-α), IL-8, IL-6, MMP-13, MMP-9, MMP-3, and prostaglandin E2 (PGE2) in chondrocyte were determined by enzyme-linked immunosorbernt assay. Bioinformatics, dual luciferin gene reporting, RNA pulldown, and Northern blot were used to determine the interaction between LINC01534 and miR-140-5p. RESULTS The results showed that LINC01534 was upregulated in both OA cartilage tissue and OA chondrocyte model. In addition, silencing LINC01534 significantly alleviated the inhibitory effect of IL-1β on expressions of aggrecan and collagen II in chondrocytes, and significantly downregulated the expression of matrix metalloproteinases in IL-1β-induced chondrocytes. Meanwhile, silencing LINC01534 also significantly inhibited the productions of proinflammatory factors NO, PGE2, TNF-α, IL-6, and IL-8 in the IL-1β-induced chondrocytes. Furthermore, miR-140-5p was confirmed to be a direct target of LINC01534. More importantly, inhibition of miR-140-5p significantly reversed the inhibitory effect of silencing LINC01534 on abnormal matrix degradation in the IL-1β-induced chondrocyte model of OA. CONCLUSION Therefore, LINC01534 could promote the abnormal matrix degradation and inflammatory response of OA chondrocytes through the targeted binding of miR-140-5p.
Collapse
Affiliation(s)
- Wei wei
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Shaoxuan He
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Zhihua Wang
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Junjie Dong
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Dong Xiang
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Yunxuan Li
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Lirong Ren
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Nannan Kou
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| | - Jia Lv
- Department of Traumatology, The Second
Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province,
People’s Republic of China
| |
Collapse
|
18
|
Farooq M, Khan AW, Kim MS, Choi S. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021; 10:cells10113242. [PMID: 34831463 PMCID: PMC8622657 DOI: 10.3390/cells10113242] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a large family of secretory molecules that act through tyrosine kinase receptors known as FGF receptors. They play crucial roles in a wide variety of cellular functions, including cell proliferation, survival, metabolism, morphogenesis, and differentiation, as well as in tissue repair and regeneration. The signaling pathways regulated by FGFs include RAS/mitogen-activated protein kinase (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)–protein kinase B (AKT), phospholipase C gamma (PLCγ), and signal transducer and activator of transcription (STAT). To date, 22 FGFs have been discovered, involved in different functions in the body. Several FGFs directly or indirectly interfere with repair during tissue regeneration, in addition to their critical functions in the maintenance of pluripotency and dedifferentiation of stem cells. In this review, we summarize the roles of FGFs in diverse cellular processes and shed light on the importance of FGF signaling in mechanisms of tissue repair and regeneration.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
- Correspondence:
| |
Collapse
|
19
|
Ren C, Jin J, Hu W, Chen Q, Yang J, Wu Y, Zhou Y, Sun L, Gao W, Zhang X, Tian N. Betulin Alleviates the Inflammatory Response in Mouse Chondrocytes and Ameliorates Osteoarthritis via AKT/Nrf2/HO-1/NF-κB Axis. Front Pharmacol 2021; 12:754038. [PMID: 34721040 PMCID: PMC8548689 DOI: 10.3389/fphar.2021.754038] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/06/2021] [Indexed: 12/02/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease featuring the degeneration, destruction, and ossification of cartilage. Inflammation which may facilitate OA occurrence and development is considered as the main pathological factor. Betulin, a natural product extracted from birch bark, has been commonly used for inflammation treatment; however, its role in OA remains unclear. This study is aimed to explore whether betulin can suppress IL-1β–induced inflammation in chondrocytes and alleviate OA in vitro and in vivo. In in vitro studies, the generation of pro-inflammatory factors, such as interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), prostaglandin E2 (PGE2), and nitric oxide (NO), was assessed using the enzyme-linked immunosorbent assay (ELISA) and Griess reaction. As revealed by results, betulin inhibited the expression of pro-inflammatory mediators. In addition, the protein expressions of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), matrix metalloproteinase (MMP-13), thrombospondin motifs 5 (ADAMTS5), Collagen II, and Aggrecan were quantified using Western blot analysis. We found that betulin could inhibit the generation of COX-2 and iNOS induced by IL-1β, indicating that betulin has anti-inflammatory effects in chondrocytes. Furthermore, betulin downregulates the expression of MMP-13 and ADAMTS-5 and upregulates the expression of Collagen II and Aggrecan, indicating that it can inhibit the degradation of the extracellular matrix. In mechanism, betulin activated the AKT/Nrf2 pathway and inhibited the phosphorylation of p65. In in vivo studies, administration of betulin in vivo could inhibit cartilage destruction and inflammatory progression. Therefore, these findings suggest that betulin may alleviate IL-1β–induced OA via the AKT/Nrf2/HO-1/NF-κB signal axis, and betulin may be a potential drug for the treatment of OA.
Collapse
Affiliation(s)
- Chenghao Ren
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Jie Jin
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Wei Hu
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Qi Chen
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Jian Yang
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yaosen Wu
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yifei Zhou
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Liaojun Sun
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Weiyang Gao
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Xiaolei Zhang
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| |
Collapse
|
20
|
Sumsuzzman DM, Choi J, Khan ZA, Kamenos G, Hong Y. Melatonin Maintains Anabolic-Catabolic Equilibrium and Regulates Circadian Rhythm During Osteoarthritis Development in Animal Models: A Systematic Review and Meta-analysis. Front Pharmacol 2021; 12:714974. [PMID: 34603028 PMCID: PMC8484877 DOI: 10.3389/fphar.2021.714974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022] Open
Abstract
Background: The driving force behind osteoarthritis (OA) pathogenesis is an anabolic-catabolic (a/c) imbalance. Melatonin (MT) is a key player in maintaining a/c stability and mitigates OA pathogenesis, but mechanisms underlying its effects remain poorly understood. Objectives: We performed a systematic review analyzing the experimental data that support the clinical applicability of MT in the treatment of OA pathogenesis, placing particular emphasis on the regulation of circadian rhythms and a/c balance. Methods: Major electronic databases and grey literature were used to identify related original articles. Methodological quality of all selected studies was evaluated using the SYRCLE risk of bias tool. Pooled mean differences (MDs)/standardized mean differences (SMDs) with 95% confidence intervals (CIs) were calculated to estimate the effect size. Results: Eleven trials were included in this systematic review. Compared with the control group, MT significantly decreased the levels of interleukin-1β (IL-1β; SMD = −5.45; 95% CI [−6.78, −4.12]; p < 0.00001, and histological grading scale (SMD = −3.46; 95% CI, [−5.24, −1.68]; p < 0.0001). MT significantly increased the transforming growth factor-β1 (TGF-β1; SMD = 1.17; 95% CI [0.31, 2.03]; p < 0.0007). Furthermore, core circadian clock genes Per2 and Cry1 mRNA levels were regulated by MT treatment in OA progression. Conclusion: MT may maintain a/c balance and regulate circadian rhythms during OA development. MT could be used in as adjunct with other interventions to manage pain and OA severity.
Collapse
Affiliation(s)
- Dewan Md Sumsuzzman
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae, Korea.,Biohealth Products Research Center (BPRC), Inje University, Gimhae, Korea.,Ubiquitous Healthcare and Anti-aging Research Center (u-HARC), Inje University, Gimhae, Korea.,Department of Physical Therapy, College of Healthcare Medical Science and Engineering, Gimhae, Korea
| | - Jeonghyun Choi
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, Korea.,Ubiquitous Healthcare and Anti-aging Research Center (u-HARC), Inje University, Gimhae, Korea.,Department of Physical Therapy, College of Healthcare Medical Science and Engineering, Gimhae, Korea
| | - Zeeshan Ahmad Khan
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, Korea.,Ubiquitous Healthcare and Anti-aging Research Center (u-HARC), Inje University, Gimhae, Korea.,Department of Physical Therapy, College of Healthcare Medical Science and Engineering, Gimhae, Korea
| | - George Kamenos
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae, Korea.,Biohealth Products Research Center (BPRC), Inje University, Gimhae, Korea.,Ubiquitous Healthcare and Anti-aging Research Center (u-HARC), Inje University, Gimhae, Korea
| | - Yonggeun Hong
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae, Korea.,Biohealth Products Research Center (BPRC), Inje University, Gimhae, Korea.,Ubiquitous Healthcare and Anti-aging Research Center (u-HARC), Inje University, Gimhae, Korea.,Department of Physical Therapy, College of Healthcare Medical Science and Engineering, Gimhae, Korea.,Department of Medicine, Division of Hematology/Oncology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MAUnited States
| |
Collapse
|
21
|
Xu J, Ma X. Hsa_circ_0032131 knockdown inhibits osteoarthritis progression via the miR-502-5p/PRDX3 axis. Aging (Albany NY) 2021; 13:15100-15113. [PMID: 34032607 PMCID: PMC8221332 DOI: 10.18632/aging.203073] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 04/29/2021] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is a chronic disease characterized by progressive loss of cartilage and failure of the diarthrodial joint. Circular RNAs (circRNAs) are known to participate in the pathogenesis of multiple diseases, including OA. We investigated the functions of hsa_circ_0032131, a circRNA upregulated in OA, using CHON-001 cells and an in vivo OA rat model. CHON-001 cells were treated with interleukin (IL)-1β to mimic OA in vitro. IL-1β-induced inhibition of CHON-001 growth was reversed by silencing hsa_circ_0032131. In addition, hsa_circ_0032131 knockdown reversed IL-1β-induced activation of Trx1, Cyclin D and PRDX3, whereas overexpression of PRDX3, a direct target of miR-502-5p, reversed this effect. Hsa_circ_0032131 served as a competing endogenous RNA for miR-502-5p. Moreover, knockdown of hsa_circ_0032131 attenuated OA symptoms in vivo by inactivating the STAT3 signaling pathway. Thus, silencing of hsa_circ_0032131 inhibited the progression of OA by inactivating the miR-502-5p/PRDX3/Trx1/STAT3 axis, which highlights its potential as a therapeutic target for OA.
Collapse
Affiliation(s)
- Jin Xu
- Department of Pain Treatment, Tianjin Hospital, Tianjin 300211, China
| | - Xinlong Ma
- Department of Pain Treatment, Tianjin Hospital, Tianjin 300211, China
| |
Collapse
|
22
|
Pinosylvin Shifts Macrophage Polarization to Support Resolution of Inflammation. Molecules 2021; 26:molecules26092772. [PMID: 34066748 PMCID: PMC8125806 DOI: 10.3390/molecules26092772] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
Pinosylvin is a natural stilbenoid found particularly in Scots pine. Stilbenoids are a group of phenolic compounds identified as protective agents against pathogens for many plants. Stilbenoids also possess health-promoting properties in humans; for instance, they are anti-inflammatory through their suppressing action on proinflammatory M1-type macrophage activation. Macrophages respond to environmental changes by polarizing towards proinflammatory M1 phenotype in infection and inflammatory diseases, or towards anti-inflammatory M2 phenotype, mediating resolution of inflammation and repair. In the present study, we investigated the effects of pinosylvin on M2-type macrophage activation, aiming to test the hypothesis that pinosylvin could polarize macrophages from M1 to M2 phenotype to support resolution of inflammation. We used lipopolysaccharide (LPS) to induce M1 phenotype and interleukin-4 (IL-4) to induce M2 phenotype in J774 murine and U937 human macrophages, and we measured expression of M1 and M2-markers. Interestingly, along with inhibiting the expression of M1-type markers, pinosylvin had an enhancing effect on the M2-type activation, shown as an increased expression of arginase-1 (Arg-1) and mannose receptor C type 1 (MRC1) in murine macrophages, and C-C motif chemokine ligands 17 and 26 (CCL17 and CCL26) in human macrophages. In IL-4-treated macrophages, pinosylvin enhanced PPAR-γ expression but had no effect on STAT6 phosphorylation. The results show, for the first time, that pinosylvin shifts macrophage polarization from the pro-inflammatory M1 phenotype towards M2 phenotype, supporting resolution of inflammation and repair.
Collapse
|
23
|
Nummenmaa E, Hämäläinen M, Pemmari A, Moilanen LJ, Tuure L, Nieminen RM, Moilanen T, Vuolteenaho K, Moilanen E. Transient Receptor Potential Ankyrin 1 (TRPA1) Is Involved in Upregulating Interleukin-6 Expression in Osteoarthritic Chondrocyte Models. Int J Mol Sci 2020; 22:ijms22010087. [PMID: 33374841 PMCID: PMC7794684 DOI: 10.3390/ijms22010087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022] Open
Abstract
Transient receptor potential ankyrin 1 (TRPA1) is a membrane-bound ion channel found in neurons, where it mediates nociception and neurogenic inflammation. Recently, we have discovered that TRPA1 is also expressed in human osteoarthritic (OA) chondrocytes and downregulated by the anti-inflammatory drugs aurothiomalate and dexamethasone. We have also shown TRPA1 to mediate inflammation, pain, and cartilage degeneration in experimental osteoarthritis. In this study, we investigated the role of TRPA1 in joint inflammation, focusing on the pro-inflammatory cytokine interleukin-6 (IL-6). We utilized cartilage/chondrocytes from wild-type (WT) and TRPA1 knockout (KO) mice, along with primary chondrocytes from OA patients. The results show that TRPA1 regulates the synthesis of the OA-driving inflammatory cytokine IL-6 in chondrocytes. IL-6 was highly expressed in WT chondrocytes, and its expression, along with the expression of IL-6 family cytokines leukemia inhibitory factor (LIF) and IL-11, were significantly downregulated by TRPA1 deficiency. Furthermore, treatment with the TRPA1 antagonist significantly downregulated the expression of IL-6 in chondrocytes from WT mice and OA patients. The results suggest that TRPA1 is involved in the upregulation of IL-6 production in chondrocytes. These findings together with previous results on the expression and functions of TRPA1 in cellular and animal models point to the role of TRPA1 as a potential mediator and novel drug target in osteoarthritis.
Collapse
Affiliation(s)
- Elina Nummenmaa
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Antti Pemmari
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Lauri J. Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Lauri Tuure
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Riina M. Nieminen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Teemu Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
- Coxa Hospital for Joint Replacement, FI-33520 Tampere, Finland
| | - Katriina Vuolteenaho
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, FI-33014 Tampere, Finland; (E.N.); (M.H.); (A.P.); (L.J.M.); (L.T.); (R.M.N.); (T.M.); (K.V.)
- Correspondence:
| |
Collapse
|
24
|
Dou P, He Y, Yu B, Duan J. Downregulation of microRNA-29b by DNMT3B decelerates chondrocyte apoptosis and the progression of osteoarthritis via PTHLH/CDK4/RUNX2 axis. Aging (Albany NY) 2020; 13:7676-7690. [PMID: 33177241 PMCID: PMC7993672 DOI: 10.18632/aging.103778] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/27/2020] [Indexed: 12/24/2022]
Abstract
The correlation between DNA methyltransferases (DNMTs) and microRNAs (miRNAs) has been well-established, but its interaction in osteoarthritis (OA) has been barely clarified. This study aimed to analyze the relationship between DNMT3B and miR-29b as well as their implications in OA. Our results revealed that DNMT3B was downregulated while miR-29b was upregulated in OA cartilage tissues relative to normal cartilage tissues. Hypermethylation of specific CpG sites in the miR-29b promoter region induced by DNMT3B contributed to downregulation of miR-29b in OA chondrocytes. Furthermore, luciferase activity determination demonstrated that miR-29b targeted and negatively regulated the parathyroid hormone-like hormone (PTHLH). Moreover, the PTHLH upregulation induced by miR-29b methylation led to the enhancement of chondrocyte growth and suppression of their apoptosis and extracellular matrix degradation, which was achieved by the upregulation cyclin-dependent kinase 4 (CDK4) expression. Co-IP suggested that CDK4 induced ubiquitination of RUNX2, which could be enhanced by DNMT3B. In the OA mouse model induced by destabilization of the medial meniscus, overexpression of DNMT3B was observed to downregulate the expression of RUNX2 whereby preventing OA-induced loss of chondrocytes. Hence, the DNMT3B/miR-29b/PTHLH/CDK4/RUNX2 axis was found to be involved in the apoptosis of chondrocytes induced by OA, highlighting a novel mechanism responsible for OA progression.
Collapse
Affiliation(s)
- Pengcheng Dou
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha 410011, P.R. China
| | - Yu He
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha 410011, P.R. China
| | - Bo Yu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, P.R. China
| | - Juan Duan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, P.R. China
| |
Collapse
|
25
|
Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, Chen H, Sun X, Feng JQ, Qi H, Chen L. FGF/FGFR signaling in health and disease. Signal Transduct Target Ther 2020; 5:181. [PMID: 32879300 PMCID: PMC7468161 DOI: 10.1038/s41392-020-00222-7] [Citation(s) in RCA: 464] [Impact Index Per Article: 92.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
Growing evidences suggest that the fibroblast growth factor/FGF receptor (FGF/FGFR) signaling has crucial roles in a multitude of processes during embryonic development and adult homeostasis by regulating cellular lineage commitment, differentiation, proliferation, and apoptosis of various types of cells. In this review, we provide a comprehensive overview of the current understanding of FGF signaling and its roles in organ development, injury repair, and the pathophysiology of spectrum of diseases, which is a consequence of FGF signaling dysregulation, including cancers and chronic kidney disease (CKD). In this context, the agonists and antagonists for FGF-FGFRs might have therapeutic benefits in multiple systems.
Collapse
Affiliation(s)
- Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Huabing Qi
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
26
|
Fibroblast growth factor signalling in osteoarthritis and cartilage repair. Nat Rev Rheumatol 2020; 16:547-564. [PMID: 32807927 DOI: 10.1038/s41584-020-0469-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
Abstract
Regulated fibroblast growth factor (FGF) signalling is a prerequisite for the correct development and homeostasis of articular cartilage, as evidenced by the fact that aberrant FGF signalling contributes to the maldevelopment of joints and to the onset and progression of osteoarthritis. Of the four FGF receptors (FGFRs 1-4), FGFR1 and FGFR3 are strongly implicated in osteoarthritis, and FGFR1 antagonists, as well as agonists of FGFR3, have shown therapeutic efficacy in mouse models of spontaneous and surgically induced osteoarthritis. FGF18, a high affinity ligand for FGFR3, is the only FGF-based drug currently in clinical trials for osteoarthritis. This Review covers the latest advances in our understanding of the molecular mechanisms that regulate FGF signalling during normal joint development and in the pathogenesis of osteoarthritis. Strategies for FGF signalling-based treatment of osteoarthritis and for cartilage repair in animal models and clinical trials are also introduced. An improved understanding of FGF signalling from a structural biology perspective, and of its roles in skeletal development and diseases, could unlock new avenues for discovery of modulators of FGF signalling that can slow or stop the progression of osteoarthritis.
Collapse
|
27
|
Chen Z, Li C, Qian YH, Fu Y, Feng ZM. Enhancement of autophagy flux by isopsoralen ameliorates interleukin-1β-stimulated apoptosis in rat chondrocytes. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2020; 22:179-192. [PMID: 30621446 DOI: 10.1080/10286020.2018.1537265] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/10/2018] [Indexed: 06/09/2023]
Abstract
Chondrocyte apoptosis contributes to the pathogenesis of cartilage degeneration in osteoarthritis (OA). We found that isopsoralen pretreatment significantly reversed the increase in DNA fragmentation and apoptosis rate, and significantly decreased the caspase-3 activity and PARP cleavage in IL-1β-treated chondrocytes. Isopsoralen pretreatment markedly inhibited disruption of matrix proteins. Moreover, the expressions of LC3-II and LAMP-1 were markedly increased but the expression of p62/SQSTM1 was remarkably decreased by isopsoralen pretreatment. Importantly, the protective effects of isopsoralen against IL-1β were blocked by pretreatment with autophagy inhibitor 3-MA and bafilomycin A1. These results suggest that isopsoralen ameliorates chondrocyte apoptosis by promoting autophagy flux.[Formula: see text].
Collapse
Affiliation(s)
- Zhi Chen
- Department of Critical Care Medicine, People's Hospital of Jiangxi Province, Nanchang 330006, China
| | - Chen Li
- Department of Orthopedics, People's Hospital of Jiangxi Province, Nanchang 330006, China
| | - Yi-Hong Qian
- Department of Anesthesiology, People's Hospital of Jiangxi Province, Nanchang 330006, China
| | - Yang Fu
- Department of Orthopedics, People's Hospital of Jiangxi Province, Nanchang 330006, China
| | - Zi-Ming Feng
- Department of Orthopedics, People's Hospital of Jiangxi Province, Nanchang 330006, China
| |
Collapse
|
28
|
Wang X, Fan J, Ding X, Sun Y, Cui Z, Liu W. Tanshinone I Inhibits IL-1β-Induced Apoptosis, Inflammation And Extracellular Matrix Degradation In Chondrocytes CHON-001 Cells And Attenuates Murine Osteoarthritis. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3559-3568. [PMID: 31686786 PMCID: PMC6800556 DOI: 10.2147/dddt.s216596] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 09/06/2019] [Indexed: 12/16/2022]
Abstract
Background Osteoarthritis (OA) is a prevalent degenerative joint disease, which was characterized by inflammation and cartilage degradation. Accumulating evidence has demonstrated that Tanshinone I has an anti-inflammatory effect in various diseases. However, the efficacy of Tanshinone I as an anti-inflammatory agent in OA remains unclear. This study aimed to explore the role of Tanshinone I on OA both in vitro and in vivo. Methods CHON-001 cells were treated with IL-1β (10 ng/mL) for 72 hrs to induce OA model in vitro. Meanwhile, CHON-001 cells were pre-treated with 20 μM Tanshinone I for 24 hrs and then stimulated with IL-1β (10 ng/mL) for 72 hrs. CCK-8, immunofluorescence and flow cytometry assays were used to detect the viability, proliferation and apoptosis in CHON-001 cells, respectively. Western blotting assay was used to detect the levels of collagen II, aggrecan, MMP-13, cleaved caspase 1, Gasdermin D, SOX11 and p-NF-κB in CHON-001 cells. In addition, the mouse model of OA was built by anterior cruciate ligament transection (ACLT) in the right knee. Meanwhile, the mice were administrated with 10 or 30 mg/kg Tanshinone I for 8 weeks. Safranin-O/Fast Green staining was used to assess cartilage destruction in a mouse model of OA. Results In this study, IL-1β significantly induced apoptosis, extracellular matrix degradation and inflammatory response in CHON-001 cells. Tanshinone I significantly inhibited IL-1β-induced apoptosis in CHON-001 cells. In addition, the IL-1β-induced collagen II, aggrecan degradation, SOX11 downregulation, and MMP-13 and p-NF-κB upregulation in CHON-001 cells were notably reversed by Tanshinone I treatment. Moreover, Tanshinone I alleviated cartilage destruction and synovitis and reduced OARSI scores and subchondral bone thickness in a mouse model of OA. Conclusion Our findings showed that Tanshinone I could alleviate the progression of OA in vitro and in vivo. These results demonstrated that Tanshinone I might be regarded as a promising therapeutic agent for the treatment of OA.
Collapse
Affiliation(s)
- Xipeng Wang
- Department of Orthopaedic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, People's Republic of China
| | - Jianbo Fan
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Xiaomin Ding
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Yuyu Sun
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Zhiming Cui
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| | - Wei Liu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, People's Republic of China
| |
Collapse
|
29
|
Bruczko-Goralewska M, Romanowicz L, Bączyk J, Wolańska M, Sobolewski K, Kowalewski R. Peptide growth factors and their receptors in the vein wall. J Investig Med 2019; 67:1149-1154. [PMID: 31427387 DOI: 10.1136/jim-2019-001075] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/03/2019] [Accepted: 07/06/2019] [Indexed: 12/29/2022]
Abstract
The varicose vein wall remodeling is a very complex process, which is controlled by numerous factors, including peptide growth factors. The aim of the study was to assess a/b FGF, IGF-1, TGF-β1, VEGF-A and their receptors in the vein wall. Varicose vein samples were taken from 24 patients undergoing varicose vein surgery. The control material consisted of vein specimens collected from 12 patients with chronic limb ischemia. Contents of aFGF, bFGF, IGF-I, TGF-β1, VEGF, IGF-1R, VEGF R1 and VEGF R2 were assessed with ELISA method. Protein expression of FGF R1 and TGF-β RII were evaluated with western blot. Increased contents of aFGF, IGF-1 and VEGF-A were found in varicose veins in comparison with normal ones (p<0.05). In contrast, a significant decrease in TGF-β content was demonstrated in varicose veins (p<0.05). Furthermore, there was no difference in bFGF content in both groups (p>0.05). IGF-1 R content was significantly increased in varicose veins (p<0.05). There was no difference in VEGF R1 content between varicose and normal veins (p>0.05), whereas VEGF R2 content was significantly increased in varicose veins (p<0.05). Western blot demonstrated increased expression of TGF-β RII in varicose veins (p<0.05) and similar expression of FGF R1 in both groups (p>0.05). Demonstrated changes in peptide growth factors and their receptors may disturb metabolism of extracellular matrix in the varicose vein wall and contribute to the development of the disease to its more advanced stages.
Collapse
Affiliation(s)
| | - Lech Romanowicz
- Department of Medical Biochemistry, Medical University of Bialystok, Bialystok, Poland
| | - Justyna Bączyk
- Department of Medical Biochemistry, Medical University of Bialystok, Bialystok, Poland
| | - Małgorzata Wolańska
- Department of Medical Biochemistry, Medical University of Bialystok, Bialystok, Poland
| | - Krzysztof Sobolewski
- Department of Medical Biochemistry, Medical University of Bialystok, Bialystok, Poland
| | - Radosław Kowalewski
- Department of General, Vascular and Oncological Surgery, Międzylesie Interdisciplinary Hospital, Warsaw, Poland
| |
Collapse
|
30
|
Wu Y, Lin Z, Yan Z, Wang Z, Fu X, Yu K. Sinomenine contributes to the inhibition of the inflammatory response and the improvement of osteoarthritis in mouse-cartilage cells by acting on the Nrf2/HO-1 and NF-κB signaling pathways. Int Immunopharmacol 2019; 75:105715. [PMID: 31310911 DOI: 10.1016/j.intimp.2019.105715] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022]
Abstract
Pathological changes, such as articular cartilage degeneration, destruction, and hyperosteogeny, are regarded as the main features of osteoarthritis (OA). Sinomenine (SIN) is a monomeric component purified from the plant Sinomenium acutum which has been found to have anti-inflammatory effects, however, the mechanism of action of SIN on OA is not clear. In this study, we evaluated whether SIN could regulate the inflammatory response induced by interleukin (IL)-1β and improve outcomes in the instability model of OA (medial meniscus mice (DMM)) by acting on the Nrf2/HO-1 and NF-κ B signaling pathways in chondrocytes. From our experiments, which include Griess reaction, ELISA, Western blot, and immunofluorescence, we found that SIN not only down-regulated the expression of pro-inflammatory factors induced by IL-1β, including; inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nitricoxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), but also decreased the production of IL-1β-induced cartilage matrix catabolic enzymes including; ADAMTS-5 and MMPs, in mouse chondrocytes. In addition, the degradation of aggrecan and type II collagen protein in the extracellular matrix (ECM) stimulated by IL-1β was reversed. Most importantly, we have revealed for the first time that in OA, SIN inhibited the inflammatory response and ECM degradation by activating the Nrf2/HO-1 signaling pathways and inhibiting NF-κB activity in mouse-cartilage cells. In in vivo experiments, SIN treatment helped to improve the cartilage destruction in OA model mice. In conclusion, this study has demonstrated that SIN inhibits the IL-1β-induced inflammatory response and cartilage destruction by activating the Nrf2/HO-1 signaling pathway and inhibiting the NF-κB signaling pathway in mouse chondrocytes, suggesting a new use for SIN in the treatment of OA.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, Zhejiang 325000, PR China; The Second School of Medicine, WenZhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Zeng Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, Zhejiang 325000, PR China; The Second School of Medicine, WenZhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Zijian Yan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, Zhejiang 325000, PR China; The Second School of Medicine, WenZhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Zhanghong Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; The Second School of Medicine, WenZhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Xin Fu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China; Zhejiang Provincial Key Laboratory of Orthpaedics, Wenzhou, Zhejiang 325000, PR China; The Second School of Medicine, WenZhou Medical University, Wenzhou, Zhejiang 325000, PR China
| | - Kehe Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, PR China.
| |
Collapse
|
31
|
Barbon S, Stocco E, Macchi V, Contran M, Grandi F, Borean A, Parnigotto PP, Porzionato A, De Caro R. Platelet-Rich Fibrin Scaffolds for Cartilage and Tendon Regenerative Medicine: From Bench to Bedside. Int J Mol Sci 2019; 20:ijms20071701. [PMID: 30959772 PMCID: PMC6479320 DOI: 10.3390/ijms20071701] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 12/22/2022] Open
Abstract
Nowadays, research in Tissue Engineering and Regenerative Medicine is focusing on the identification of instructive scaffolds to address the requirements of both clinicians and patients to achieve prompt and adequate healing in case of injury. Among biomaterials, hemocomponents, and in particular Platelet-rich Fibrin matrices, have aroused widespread interest, acting as delivery platforms for growth factors, cytokines and immune/stem-like cells for immunomodulation; their autologous origin and ready availability are also noteworthy aspects, as safety- and cost-related factors and practical aspects make it possible to shorten surgical interventions. In fact, several authors have focused on the use of Platelet-rich Fibrin in cartilage and tendon tissue engineering, reporting an increasing number of in vitro, pre-clinical and clinical studies. This narrative review attempts to compare the relevant advances in the field, with particular reference being made to the regenerative role of platelet-derived growth factors, as well as the main pre-clinical and clinical research on Platelet-rich Fibrin in chondrogenesis and tenogenesis, thereby providing a basis for critical revision of the topic.
Collapse
Affiliation(s)
- Silvia Barbon
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Elena Stocco
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Veronica Macchi
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Martina Contran
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
| | - Francesca Grandi
- Complex Operative Unit-Pediatric Surgery, Hospital of Bolzano, Via L. Böhler 5, 39100 Bolzano, Italy.
| | - Alessio Borean
- Department of Immunohematology and Transfusion Medicine, San Martino Hospital, 32100 Belluno, Italy.
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (T.E.S.) Onlus, 35131 Padua, Italy.
| | - Andrea Porzionato
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| | - Raffaele De Caro
- Department of Neuroscience, Section of Human Anatomy, University of Padova, Via A. Gabelli 65, 35121 Padova, Italy.
- LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Veneto Region, Via N. Giustiniani 2, 35128 Padova, Italy.
| |
Collapse
|
32
|
Duan ZX, Huang P, Tu C, Liu Q, Li SQ, Long ZL, Li ZH. MicroRNA-15a-5p Regulates the Development of Osteoarthritis by Targeting PTHrP in Chondrocytes. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3904923. [PMID: 30949498 PMCID: PMC6425345 DOI: 10.1155/2019/3904923] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 02/20/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS A growing body of research has demonstrated that the degeneration of chondrocytes is the primary cause of osteoarthritis (OA). Parathyroid hormone-related protein (PTHrP) can alleviate the degeneration of chondrocytes via promotion of chondrocyte proliferation and inhibition of terminal differentiation, but the underlying mechanism remains unknown. This study aimed to identify the microRNAs (miRNAs) that may target PTHrP and regulate the proliferation and terminal differentiation of chondrocytes. METHODS Bioinformatic analysis was used to predict which miRNAs target PTHrP. We collected human knee cartilage specimens to acquire the primary chondrocytes, which we then used to test the expression and function of the targeted miRNAs. To explore the effects of miR-15a-5p on the putative binding sites, specific mimics or inhibitors were transfected into the chondrocytes. Furthermore, a dual-luciferase reporter gene assay and chondrocyte degeneration-related factors were used to verify the possible mechanism. RESULTS The expression of PTHrP was upregulated in the OA chondrocytes, whilst miR-15a-5p was downregulated in the OA chondrocytes. A negative correlation was observed between PTHrP and miR-15a-5p. The knockdown of miR-15a-5p promoted the growth of chondrocytes and inhibited calcium deposition, whilst overexpression of miR-15a-5p reversed this trend. The effect of miR-15a-5p overexpression was neutralised by PTHrP. Dual-luciferase reporter assays revealed that PTHrP can be used as a novel targeting molecule for miR-15a-5p. CONCLUSIONS miR-15a-5p promotes the degeneration of chondrocytes by targeting PTHrP and, in addition to helping us understand the development of OA, may be a potential biomarker of OA.
Collapse
Affiliation(s)
- Zhi-xi Duan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha 410011, China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha 410008, China
| | - Chao Tu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha 410011, China
| | - Qing Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha 410011, China
| | - Shuang-qing Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha 410011, China
| | - Ze-ling Long
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha 410011, China
| | - Zhi-hong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha 410011, China
| |
Collapse
|
33
|
Zhang W, Wu J, Zhang F, Dou X, Ma A, Zhang X, Shao H, Zhao S, Ling P, Liu F, Han G. Lower range of molecular weight of xanthan gum inhibits apoptosis of chondrocytes through MAPK signaling pathways. Int J Biol Macromol 2019; 130:79-87. [PMID: 30659877 DOI: 10.1016/j.ijbiomac.2019.01.071] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/14/2019] [Accepted: 01/16/2019] [Indexed: 01/04/2023]
Abstract
LRWXG has previously been reported to have a protective effect on chondrocytes, preventing apoptosis induced by oxidative stress. In this study, we were aimed at determining whether LRWXG exerts its anti-apoptotic activity through the MAPK signaling pathways in chondrocytes. Our results show that, at the cellular level, apoptosis of chondrocytes in the groups treated by LRWXG decreases compared with groups treated by inhibitors alone and model group under conditions of oxidative stress in a dose-dependent manner. Mechanistically at the molecular level, LRWXG regulates the MAPK pathway induced by oxidative stress: The levels of phosphorylation of JNK and p38 proteins in the groups treated by LRWXG are lower than model group, while compared with corresponding groups of inhibitors, there are no significant difference; For other related proteins, LRWXG reduces the levels of the apoptosis-related proteins BAX and cleaved caspase-3, and increases the level of anti-apoptotic protein BCL2. In addition, LRWXG can significantly reduce the levels of inflammatory-related factors such as COX2, PEG2, TNFα and IL1β, and inhibits the expression of MMPs, increasing the content of type II collagen. The results of this research strongly suggest that LRWXG exerts its anti-apoptotic activity via regulating the MAPK signaling pathways in vitro.
Collapse
Affiliation(s)
- Wei Zhang
- Jinzhou Medical University, Jinzhou 121001, China; The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China; Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Jixu Wu
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Fangfang Zhang
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Xixi Dou
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Aibin Ma
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Xiaogang Zhang
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Huarong Shao
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China
| | - Shuo Zhao
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Peixue Ling
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China; School of Pharmaceutical Sciences, Shandong University, Jinan 250101, China
| | - Fei Liu
- Shandong Academy of Pharmaceutical Science, Key Laboratory of Biopharmaceuticals, Engineering Laboratory of Polysaccharide Drugs, National-Local Joint Engineering Laboratory of Polysaccharide Drugs, Jinan 250101, China.
| | - Guanying Han
- Jinzhou Medical University, Jinzhou 121001, China; The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China.
| |
Collapse
|
34
|
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
|
35
|
Xu L, Zhang L, Zhang H, Yang Z, Qi L, Wang Y, Ren S. The participation of fibroblast growth factor 23 (FGF23) in the progression of osteoporosis via JAK/STAT pathway. J Cell Biochem 2018; 119:3819-3828. [PMID: 28782829 DOI: 10.1002/jcb.26332] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/04/2017] [Indexed: 01/09/2023]
Abstract
Osteoporosis (OP) is a major skeletal disorder for the old man. The fibroblast growth factor 23 (FGF23) is a phosphaturic hormone produced by osteoblasts and osteocytes. However, the regulatory mechanisms of FGF23 in the progression of osteoporosis remain poorly understood. This study aims to explore the downstream regulating pathway of FGF23 in postmenopausal osteoporosis. The rat model of osteoporosis was established through ovariectomy (OVX). The investigation demonstrated that the serum levels of FGF23 and the phosphorylation levels of JAK2, STAT1, and STAT3 were up-regulated in the OVX + NVP-BGJ398 group while were down-regulated in the OVX + Anti-FGF23 group than that in the OVX group. Moreover, the JAK2/STAT1/3 inhibitor, AG490 promoted the OVX-induced increase in the osteocalcin, ALP, BALP, TRAP, and CTX-I levels. Besides, AG490 enhanced cartilage lesions and increased TUNEL-positive chondrocytes in the OVX group. In addition, higher protein expression of MMP-1 and MMP-13 and lower expression of COX-II were observed in the OVX + AG490 group than that in the OVX group. Our findings suggested that FGF23 was involved in the progression of osteoporosis via the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Lijun Xu
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lixia Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huijuan Zhang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zaigang Yang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Qi
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yurong Wang
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuxin Ren
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
36
|
Jeyakumar V, Niculescu-Morzsa E, Bauer C, Lacza Z, Nehrer S. Platelet-Rich Plasma Supports Proliferation and Redifferentiation of Chondrocytes during In Vitro Expansion. Front Bioeng Biotechnol 2017; 5:75. [PMID: 29270404 PMCID: PMC5723650 DOI: 10.3389/fbioe.2017.00075] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/23/2017] [Indexed: 01/05/2023] Open
Abstract
Articular cartilage regeneration is insufficient to restore sports injuries or defects that can occur from trauma. Treatment options for cartilage repair include autologous chondrocyte implantation (ACI) by isolation, expansion, and reimplantation of healthy donor chondrocytes. Chondrocyte expansion onto 2D substrates leads to dedifferentiation and loss of the cellular phenotype. We aimed to overcome the state of dedifferentiation by biochemical stimuli with platelet derivatives such as platelet-rich plasma (PRP) and hyperacute serum (HAS) to achieve sufficient cell numbers in combination with variable oxygen tension. Human articular chondrocytes from osteoarthritic (OA) cartilage chondrocytes were switched from 10% FCS supplementation to either 10% PRP or 10% HAS after initial passaging for further experiments under normoxic (20% O2) or hypoxic (1% O2) conditions. An XTT assay measured the effect of PRP or HAS on the cell proliferation at 3, 6, and 9 days. The chondrogenic redifferentiation potential of dedifferentiated chondrocytes was determined with reverse transcriptase quantitative real-time PCR for markers of expression for type II collagen (COL2A1), type I collagen (COL1A1), and matrix metalloproteinases MMP3, matrix metalloproteinase 13 (MMP13) at 24 and 72 h. Measured protein levels of 100% PRP or HAS by multiplex quantification revealed basic fibroblast growth factor, G-CSF, and PDGF were significantly higher in PRP than in HAS (p < 0.05) but LEPTIN levels did not differ. The quantified protein levels did not differ when isolated from same donors at a different time. Chondrocyte proliferation indicated that supplementation of 10% HAS enhanced the proliferation rate compared to 10% PRP or 10% FCS at 6 and 9 days significantly (p < 0.05). mRNA levels for expression of COL1A1 were significantly downregulated (p < 0.05) when cultured with 10% PRP than 10% HAS or 10% FCS under normoxic/hypoxic conditions. COL2A1 was significantly upregulated (p < 0.05) in PRP than 10% HAS or 10% FCS. MMP3 expression was downregulated after 72 h under all conditions. MMP13 was upregulated with 10% PRP at both 24 and 72 h but significantly downregulated under hypoxia (1% O2) for all circumstances. While HAS has its effect on chondrocyte proliferation, PRP enhances both proliferation and redifferentiation of dedifferentiated chondrocytes. PRP can replace standard usage of FCS for chondrogenic priming and expansion as implications for clinical use such as ACI procedures.
Collapse
Affiliation(s)
- Vivek Jeyakumar
- Centre for Regenerative Medicine and Orthopedics, Danube University Krems, Krems an der Donau, Austria
| | - Eugenia Niculescu-Morzsa
- Centre for Regenerative Medicine and Orthopedics, Danube University Krems, Krems an der Donau, Austria
| | - Christoph Bauer
- Centre for Regenerative Medicine and Orthopedics, Danube University Krems, Krems an der Donau, Austria
| | | | - Stefan Nehrer
- Centre for Regenerative Medicine and Orthopedics, Danube University Krems, Krems an der Donau, Austria
| |
Collapse
|
37
|
Platelet-rich plasma induces post-natal maturation of immature articular cartilage and correlates with LOXL1 activation. Sci Rep 2017. [PMID: 28623328 PMCID: PMC5473810 DOI: 10.1038/s41598-017-02297-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Platelet-rich plasma (PRP) is used to stimulate the repair of acute and chronic cartilage damage even though there is no definitive evidence of how this is achieved. Chondrocytes in injured and diseased situations frequently re-express phenotypic biomarkers of immature cartilage so tissue maturation is a potential pathway for restoration of normal structure and function. We used an in vitro model of growth factor-induced maturation to perform a comparative study in order to determine whether PRP can also induce this specific form of remodeling that is characterised by increased cellular proliferation and tissue stiffness. Gene expression patterns specific for maturation were mimicked in PRP treated cartilage, with chondromodulin, collagen types II/X downregulated, deiodinase II and netrin-1 upregulated. PRP increased cartilage surface cell density 1.5-fold (P < 0.05), confirmed by bromodeoxyuridine incorporation and proportionate increases in proliferating cell nuclear antigen gene expression. Atomic force microscopy analysis of PRP and growth factor treated cartilage gave a 5-fold increase in stiffness correlating with a 10-fold upregulation of lysyl oxidase like-1 gene expression (P < 0.001). These data show PRP induces key aspects of post-natal maturation in immature cartilage and provides the basis to evaluate a new biological rationale for its activity when used clinically to initiate joint repair.
Collapse
|
38
|
T140 blocks the SDF-1/CXCR4 signaling pathway and prevents cartilage degeneration in an osteoarthritis disease model. PLoS One 2017; 12:e0176048. [PMID: 28426786 PMCID: PMC5398617 DOI: 10.1371/journal.pone.0176048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/04/2017] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is one of the most common diseases affecting older people; however, there remains no effective targeted drug to combat OA. The aims of this study were (1) to explore the effect of T140 in regulating degeneration of articular cartilage in vivo by targeted blocking of the SDF-1/CXCR4 signaling pathway, and (2) to provide experimental evidence for the development of a novel OA-targeted pharmacotherapy. Thirty-six healthy Hartley guinea pigs were randomly divided into three groups: a T140-treated group (n = 12), a phosphate buffer saline control group (n = 12) and an untreated control group (n = 12). At 2, 4, 6, 8, 10 and 12 weeks of treatment, SDF-1 in serum was quantified by enzyme-linked immunosorbent assay. At 12 weeks of treatment, the cartilage from knee tibial plateau in the knee joint was collected for H&E, Safranin-O staining and Mankin grading; measurement for mRNA levels of matrix metalloproteinases (MMP-3, MMP-9 and MMP-13), aggrecan (ACAN) and collagen II (Col II) using RT-PCR; and measurement for Col II protein levels by western blot. Results showed that SDF-1 in serum increased in the T140 group and increased in the control groups. H&E and Safranin-O staining revealed less cartilage loss in T140-treated animals compared to controls. The mRNA levels of MMP-3, MMP-9 and MMP-13 in cartilage were much lower in the T140 group than other groups, but mRNA levels of ACAN and Col II in cartilage were higher in the T140-treated group. Col II protein levels in the T140 group and control groups were different. T140 can downregulate the expression of matrix-degrading enzyme and lessen the degeneration of cartilage by blocking the SDF-1/CRCR4 signaling pathway in vivo. This mechanism may present a pharmacological target for the treatment of OA.
Collapse
|
39
|
Passipieri JA, Baker HB, Siriwardane M, Ellenburg MD, Vadhavkar M, Saul JM, Tomblyn S, Burnett L, Christ GJ. Keratin Hydrogel Enhances In Vivo Skeletal Muscle Function in a Rat Model of Volumetric Muscle Loss. Tissue Eng Part A 2017; 23:556-571. [PMID: 28169594 DOI: 10.1089/ten.tea.2016.0458] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Volumetric muscle loss (VML) injuries exceed the considerable intrinsic regenerative capacity of skeletal muscle, resulting in permanent functional and cosmetic deficits. VML and VML-like injuries occur in military and civilian populations, due to trauma and surgery as well as due to a host of congenital and acquired diseases/syndromes. Current therapeutic options are limited, and new approaches are needed for a more complete functional regeneration of muscle. A potential solution is human hair-derived keratin (KN) biomaterials that may have significant potential for regenerative therapy. The goal of these studies was to evaluate the utility of keratin hydrogel formulations as a cell and/or growth factor delivery vehicle for functional muscle regeneration in a surgically created VML injury in the rat tibialis anterior (TA) muscle. VML injuries were treated with KN hydrogels in the absence and presence of skeletal muscle progenitor cells (MPCs), and/or insulin-like growth factor 1 (IGF-1), and/or basic fibroblast growth factor (bFGF). Controls included VML injuries with no repair (NR), and implantation of bladder acellular matrix (BAM, without cells). Initial studies conducted 8 weeks post-VML injury indicated that application of keratin hydrogels with growth factors (KN, KN+IGF-1, KN+bFGF, and KN+IGF-1+bFGF, n = 8 each) enabled a significantly greater functional recovery than NR (n = 7), BAM (n = 8), or the addition of MPCs to the keratin hydrogel (KN+MPC, KN+MPC+IGF-1, KN+MPC+bFGF, and KN+MPC+IGF-1+bFGF, n = 8 each) (p < 0.05). A second series of studies examined functional recovery for as many as 12 weeks post-VML injury after application of keratin hydrogels in the absence of cells. A significant time-dependent increase in functional recovery of the KN, KN+bFGF, and KN+IGF+bFGF groups was observed, relative to NR and BAM implantation, achieving as much as 90% of the maximum possible functional recovery. Histological findings from harvested tissue at 12 weeks post-VML injury documented significant increases in neo-muscle tissue formation in all keratin treatment groups as well as diminished fibrosis, in comparison to both BAM and NR. In conclusion, keratin hydrogel implantation promoted statistically significant and physiologically relevant improvements in functional outcomes post-VML injury to the rodent TA muscle.
Collapse
Affiliation(s)
- J A Passipieri
- 1 Biomedical Engineering Department, University of Virginia , Charlottesville, Virginia.,2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - H B Baker
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina.,3 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | - Mevan Siriwardane
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | | | - Manasi Vadhavkar
- 2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina
| | - Justin M Saul
- 5 Department of Chemical, Paper and Biomedical Engineering, Miami University , Oxford, Ohio
| | - Seth Tomblyn
- 4 KeraNetics, LLC , Winston-Salem, North Carolina
| | - Luke Burnett
- 4 KeraNetics, LLC , Winston-Salem, North Carolina
| | - George J Christ
- 1 Biomedical Engineering Department, University of Virginia , Charlottesville, Virginia.,2 Wake Forest Institute for Regenerative Medicine, Wake Forest University , Winston-Salem, North Carolina.,6 Orthopaedics Department, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
40
|
Huang Y, Wan G, Tao J. C1q/TNF-related protein-3 exerts the chondroprotective effects in IL-1β-treated SW1353 cells by regulating the FGFR1 signaling. Biomed Pharmacother 2017; 85:41-46. [DOI: 10.1016/j.biopha.2016.11.128] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/22/2016] [Accepted: 11/27/2016] [Indexed: 10/20/2022] Open
|
41
|
Bianchi A, Guibert M, Cailotto F, Gasser A, Presle N, Mainard D, Netter P, Kempf H, Jouzeau JY, Reboul P. Fibroblast Growth Factor 23 drives MMP13 expression in human osteoarthritic chondrocytes in a Klotho-independent manner. Osteoarthritis Cartilage 2016; 24:1961-1969. [PMID: 27307356 DOI: 10.1016/j.joca.2016.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Fibroblast Growth Factor 23 (FGF23) may represent an attractive candidate that could participate to the osteoarthritic (OA)-induced phenotype switch of chondrocytes. To address this hypothesis, we investigated the expression of FGF23, its receptors (FGFRs) and co-receptor (Klotho) in human cartilage and studied the effects of rhFGF23 on OA chondrocytes. METHOD Gene expression or protein levels were analysed by RT-PCR and immunohistochemistry. Collagenase 3 (MMP13) activity was measured by a fluorescent assay. MAPK signalling pathways were investigated by phosphoprotein array, immunoblotting and the use of selective inhibitors. RNA silencing was performed to confirm the respective contribution of FGFR1 and Klotho. RESULTS We showed that the expression of FGF23, FGFR1 and Klotho was up-regulated at both mRNA and protein levels in OA chondrocytes when compared to healthy ones. These overexpressions were markedly elevated in the damaged regions of OA cartilage. When stimulated with rhFGF23, OA chondrocytes displayed an extended expression of FGF23 and of markers of hypertrophy such as MMP13, COL10A1, and VEGF. We demonstrated that FGF23 auto-stimulation was both FGFR1-and Klotho-dependent, whereas the expression of markers of hypertrophy was mainly dependent on FGFR1 alone. Finally, we showed that FGF23-induced MMP13 expression was strongly regulated by the MEK/ERK cascade and to a lesser extent, by the PI-3K/AKT pathway. CONCLUSION These results demonstrate that FGF23 sustains differentiation of OA chondrocytes in a Klotho-independent manner.
Collapse
Affiliation(s)
- A Bianchi
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - M Guibert
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - F Cailotto
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - A Gasser
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - N Presle
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - D Mainard
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Chirurgie Orthopédique et Traumatologique, Centre Hospitalier Universitaire, Nancy, France.
| | - P Netter
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Centre Hospitalier Universitaire, Nancy, France.
| | - H Kempf
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| | - J-Y Jouzeau
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France; Département de Pharmacologie Clinique et Toxicologie, Centre Hospitalier Universitaire, Nancy, France.
| | - P Reboul
- UMR 7365 CNRS-Université de Lorraine « Ingénierie Moléculaire et Physiopathologie Articulaire » (IMoPA), Biopôle de l'Université de Lorraine, Campus Biologie-Santé, Vandœuvre-lès-Nancy, France.
| |
Collapse
|
42
|
Nummenmaa E, Hämäläinen M, Moilanen LJ, Paukkeri EL, Nieminen RM, Moilanen T, Vuolteenaho K, Moilanen E. Transient receptor potential ankyrin 1 (TRPA1) is functionally expressed in primary human osteoarthritic chondrocytes. Arthritis Res Ther 2016; 18:185. [PMID: 27515912 PMCID: PMC4982008 DOI: 10.1186/s13075-016-1080-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/21/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transient receptor potential ankyrin 1 (TRPA1) is a membrane-associated cation channel, widely expressed in neuronal cells and involved in nociception and neurogenic inflammation. We showed recently that TRPA1 mediates cartilage degradation and joint pain in the MIA-model of osteoarthritis (OA) suggesting a hitherto unknown role for TRPA1 in OA. Therefore, we aimed to investigate whether TRPA1 is expressed and functional in human OA chondrocytes. METHODS Expression of TRPA1 in primary human OA chondrocytes was assessed by qRT-PCR and Western blot. The functionality of the TRPA1 channel was assessed by Ca(2+)-influx measurements. Production of MMP-1, MMP-3, MMP-13, IL-6, and PGE2 subsequent to TRPA1 activation was measured by immunoassay. RESULTS We show here for the first time that TRPA1 is expressed in primary human OA chondrocytes and its expression is increased following stimulation with inflammatory factors IL-1β, IL-17, LPS, and resistin. Further, the TRPA1 channel was found to be functional, as stimulation with the TRPA1 agonist AITC caused an increase in Ca(2+) influx, which was attenuated by the TRPA1 antagonist HC-030031. Genetic depletion and pharmacological inhibition of TRPA1 downregulated the production of MMP-1, MMP-3, MMP-13, IL-6, and PGE2 in osteoarthritic chondrocytes and murine cartilage, respectively. CONCLUSIONS The TRPA1 cation channel was found to be functionally expressed in primary human OA chondrocytes, which is an original finding. The presence and inflammatory and catabolic effects of TRPA1 in human OA chondrocytes propose a highly intriguing role for TRPA1 as a pathogenic factor and drug target in OA.
Collapse
Affiliation(s)
- Elina Nummenmaa
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Lauri J Moilanen
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Erja-Leena Paukkeri
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Riina M Nieminen
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Teemu Moilanen
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland.,Coxa Hospital for Joint Replacement, Tampere, Finland
| | - Katriina Vuolteenaho
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland.
| |
Collapse
|
43
|
Patel D, Gao Y, Son K, Siltanen C, Neve RM, Ferrara K, Revzin A. Microfluidic co-cultures with hydrogel-based ligand trap to study paracrine signals giving rise to cancer drug resistance. LAB ON A CHIP 2015; 15:4614-4624. [PMID: 26542093 DOI: 10.1039/c5lc00948k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Targeted cancer therapies are designed to deactivate signaling pathways used by cancer cells for survival. However, cancer cells are often able to adapt by activating alternative survival pathways, thereby acquiring drug resistance. An emerging theory is that autocrine or paracrine growth factor signaling in the cancer microenvironment represent an important mechanism of drug resistance. In the present study we wanted to examine whether paracrine interactions between groups of melanoma cells result in resistance to vemurafenib - an FDA approved drug targeting the BRAF mutation in metastatic melanoma. We used a vemurafenib-resistant melanoma model which secretes fibroblast growth factor (FGF)-2 to test our hypothesis that this is a key paracrine mediator of resistance to vemurafenib. Sensitive cells treated with media conditioned by resistant cells did not protect from the effects of vemurafenib. To query paracrine interactions further we fabricated a microfluidic co-culture device with two parallel compartments, separated by a 100 μm wide hydrogel barrier. The gel barrier prevented resorting/contact of cells while permitting paracrine cross-talk. In this microfluidic system, sensitive cells did become refractive to the effects of vemurafenib when cultured adjacent to resistant cells. Importantly, incorporation of FGF-2 capture probes into the gel barrier separating the two cell types prevented onset of resistance to vemurafenib. Microfluidic tools described here allow for more sensitive analysis of paracrine signals, may help better understand signaling in the cancer microenvironment and may enable development of more effective cancer therapies.
Collapse
Affiliation(s)
- Dipali Patel
- Department of Biomedical Engineering, One Shields Ave, University of California, Davis, CA 95618, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Gabusi E, Paolella F, Manferdini C, Gambari L, Schiavinato A, Lisignoli G. Age-independent effects of hyaluronan amide derivative and growth hormone on human osteoarthritic chondrocytes. Connect Tissue Res 2015; 56:440-51. [PMID: 26075645 DOI: 10.3109/03008207.2015.1047928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM Increased age is the most prominent risk factor for the initiation and progression of osteoarthritis (OA). The effects of human growth hormone (hGH) combined or not with hyaluronan amide derivative (HAD) were evaluated on human OA chondrocytes, to define their biological action and potentiality in OA treatment. MATERIAL AND METHODS Cell viability, metabolic activity, gene expression and factors released were tested at different time points on chondrocytes treated with different concentrations of hGH (0.01-10 μg/ml) alone or in combination with HAD (1 mg/ml). RESULTS We found that OA chondrocytes express GH receptor and that the different doses of hGH tested did not affect cell viability, metabolic activity or the expression of collagen type 2, 1, or 10 nor did it induce the release of IGF-1 or FGF-2. Conversely, hGH treatment increased the expression of hyaluronan receptor CD44. HAD combined with hGH reduced metabolic activity, IL6 release and gene expression, but not the suppressor of cytokine signaling 2 (SOCS2), which was significantly induced and translocated into the nucleus. The parameters analyzed, independently of the treatments used proportionally decreased with increasing age of the patients. CONCLUSIONS hGH only induced CD44 receptor on OA chondrocytes but did not affect other parameters, such as chondrocytic gene markers or IGF-1 or FGF-2 release. HAD reduced all the effects induced by hGH partially through a significant induction of SOCS2. These data show that GH or HAD treatment does not influence the response of the OA chondrocytes, thus the modulation of cellular response is age-independent.
Collapse
Affiliation(s)
- Elena Gabusi
- a Laboratorio RAMSES , Istituto Ortopedico Rizzoli , Bologna , Italy
| | | | - Cristina Manferdini
- a Laboratorio RAMSES , Istituto Ortopedico Rizzoli , Bologna , Italy .,b SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale , Istituto Ortopedico Rizzoli , Bologna , Italy , and
| | - Laura Gambari
- b SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale , Istituto Ortopedico Rizzoli , Bologna , Italy , and
| | | | - Gina Lisignoli
- a Laboratorio RAMSES , Istituto Ortopedico Rizzoli , Bologna , Italy .,b SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale , Istituto Ortopedico Rizzoli , Bologna , Italy , and
| |
Collapse
|