1
|
Danceanu-Zara CM, Petrovici A, Labusca L, Minuti AE, Stavila C, Plamadeala P, Tiron CE, Aniţă D, Aniţă A, Lupu N. Collection, Establishment and Assessment of Complex Human Osteocartilaginous Explants for Modeling Osteoarthritis. Biomedicines 2024; 12:2406. [PMID: 39457719 PMCID: PMC11504061 DOI: 10.3390/biomedicines12102406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
With the increasing burden of osteoarthritis worldwide, cost efficient and reliable models are needed to enable the development of innovative therapies or therapeutic interventions. Ex vivo models have been identified as valuable modalities in translational research, bridging the gap between in vitro and in vivo models. Osteocartilaginous explants from Osteoarthritis (OA) patients offer an exquisite opportunity for studying OA progression and testing novel therapies. We describe the protocol for establishing human osteocartilaginous explants with or without co-culture of homologous synovial tissue. Furthermore, a detailed protocol for the assessment of explanted tissue in terms of protein content using Western blot and immunohistochemistry is provided. Commentaries regarding the technique of choice, possible variations and expected results are inserted.
Collapse
Affiliation(s)
- Camelia-Mihaela Danceanu-Zara
- National Institute of Research and Development in Technical Physics, 700050 Iasi, Romania; (C.-M.D.-Z.); (A.E.M.); (C.S.); (N.L.)
- Transcend Center Regional Oncology Institute, 700483 Iasi, Romania;
| | - Adriana Petrovici
- Regional Center of Advanced Research for Emerging Diseases, Zoonoses and Food Safety, Faculty of Veterinary Medicine, Iași University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (A.P.); (D.A.); (A.A.)
| | - Luminita Labusca
- National Institute of Research and Development in Technical Physics, 700050 Iasi, Romania; (C.-M.D.-Z.); (A.E.M.); (C.S.); (N.L.)
- Orthopedics and Trauma Clinic, County Emergency Hospital, 700111 Iasi, Romania
| | - Anca Emanuela Minuti
- National Institute of Research and Development in Technical Physics, 700050 Iasi, Romania; (C.-M.D.-Z.); (A.E.M.); (C.S.); (N.L.)
| | - Cristina Stavila
- National Institute of Research and Development in Technical Physics, 700050 Iasi, Romania; (C.-M.D.-Z.); (A.E.M.); (C.S.); (N.L.)
| | - Petru Plamadeala
- Pathology Department, Saint Mary‘s Children Hospital, 700309 Iasi, Romania;
| | | | - Dragoş Aniţă
- Regional Center of Advanced Research for Emerging Diseases, Zoonoses and Food Safety, Faculty of Veterinary Medicine, Iași University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (A.P.); (D.A.); (A.A.)
| | - Adriana Aniţă
- Regional Center of Advanced Research for Emerging Diseases, Zoonoses and Food Safety, Faculty of Veterinary Medicine, Iași University of Life Sciences (IULS), 8 Mihail Sadoveanu Alley, 700489 Iasi, Romania; (A.P.); (D.A.); (A.A.)
| | - Nicoleta Lupu
- National Institute of Research and Development in Technical Physics, 700050 Iasi, Romania; (C.-M.D.-Z.); (A.E.M.); (C.S.); (N.L.)
| |
Collapse
|
2
|
Cramer EEA, Hermsen KCJ, Kock LM, Ito K, Hofmann S. Culture system for longitudinal monitoring of bone dynamics ex vivo. Biotechnol Bioeng 2024. [PMID: 39295202 DOI: 10.1002/bit.28848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 07/19/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024]
Abstract
To quantify and visualize both bone formation and resorption within osteochondral explants cultured ex vivo is challenging with the current analysis techniques. An approach that enables monitoring of bone remodeling dynamics is longitudinal microcomputed tomography (µCT), a non-destructive technique that relies on repeated µCT scanning and subsequent registration of consecutive scans. In this study, a two-compartment culture system suitable for osteochondral explants that allowed for µCT scanning during ex vivo culture was established. Explants were scanned repeatedly in a fixed orientation, which allowed assessment of bone remodeling due to adequate image registration. Using this method, bone formation was found to be restricted to the outer surfaces when cultured statically. To demonstrate that the culture system could capture differences in bone remodeling, explants were cultured statically and under dynamic compression as loading promotes osteogenesis. No quantitative differences between static and dynamic culture were revealed. Still, only in dynamic conditions, bone formation was visualized on trabecular surfaces located within the inner cores, suggesting enhanced bone formation towards the center of the explants upon mechanical loading. Taken together, the ex vivo culture system in combination with longitudinal µCT scanning and subsequent registration of images demonstrated potential for evaluating bone remodeling within explants.
Collapse
Affiliation(s)
- Esther E A Cramer
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Kim C J Hermsen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Linda M Kock
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- LifeTec Group BV, Eindhoven, The Netherlands
| | - Keita Ito
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Sandra Hofmann
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute of Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
3
|
Wei Q, Yu Z, Yang P, Chen X. Baicalin Maintains Articular Cartilage Homeostasis and Alleviates Osteoarthritis by Activating FOXO1. J Med Food 2024; 27:301-311. [PMID: 38377551 DOI: 10.1089/jmf.2023.k.0206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024] Open
Abstract
Baicalin has been acknowledged for its anti-inflammatory properties. However, its potential impact on osteoarthritis (OA) has not yet been explored. Therefore, our study aimed to examine the effects of Baicalin on OA, both in laboratory and animal models. To evaluate its efficacy, human chondrocytes affected by OA were treated with interleukin-1β and/or Baicalin. The effects were then assessed through viability tests using the cell counting kit-8 (CCK-8) method and flow cytometry. In addition, we analyzed the expressions of various factors such as FOXO1, autophagy, apoptosis, and cartilage synthesis and breakdown to corroborate the effects of Baicalin. We also assessed the severity of OA through analysis of tissue samples. Our findings demonstrate that Baicalin effectively suppresses inflammatory cytokines and MMP-13 levels caused by collagenase-induced osteoarthritis, while simultaneously preserving the levels of Aggrecan and Col2. Furthermore, Baicalin has been shown to enhance autophagy. Through the use of FOXO1 inhibitors, lentivirus-mediated knockdown, and chromatin immunoprecipitation, we verified that Baicalin exerts its protective effects by activating FOXO1, which binds to the Beclin-1 promoter, thereby promoting autophagy. In conclusion, our results show that Baicalin has potential as a therapeutic agent for treating OA (Clinical Trial Registration number: 2023-61).
Collapse
Affiliation(s)
- Qiang Wei
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Zhaoping Yu
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Peng Yang
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| | - Xiaohu Chen
- Department of Clinical Laboratory, The People's Hospital of Rongchang District, Chongqing, China
| |
Collapse
|
4
|
Antoniadis A, Wegrzyn J, Omoumi P, Loisay L, Hügle T, Geurts J. Elevated secretion of pro-collagen I-alpha and vascular endothelial growth factor as biomarkers of acetabular labrum degeneration and calcification in hip osteoarthritis: An explant study. J Orthop Translat 2024; 44:19-25. [PMID: 38179125 PMCID: PMC10765489 DOI: 10.1016/j.jot.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/29/2023] [Accepted: 08/23/2023] [Indexed: 01/06/2024] Open
Abstract
Background Hip osteoarthritis (OA) involves structural degeneration of different joint compartments, including femoral head cartilage, periarticular ligaments and the acetabular labrum. However, the molecular mechanisms underlying labrum degeneration in hip OA remain poorly understood. Aim To assess secretion of putative biomarkers for OA from explanted human labrum tissues under basal and inflammatory conditions and to determine whether these could differentiate between OA and calcification status compared to fracture controls. Methods Intact labrum specimens were collected from patients undergoing joint arthroplasty for primary hip OA (n = 15, mean age 70) or non-OA femoral neck fracture (n = 5, mean age 64). Tissues were dissected in equal-sized samples and explanted for one week. To mimic activation of inflammatory signaling by endogenous damage-associated molecular patterns (DAMP) tissue were stimulated with a toll-like receptor 4 (TLR4) agonist (1 μg/mL LPS). The involvement of transforming growth factor-beta (TGF-beta) signaling was evaluated by treatment with a TGF-beta type 1 receptor inhibitor (10 μM SB-505124). Secretion of aggrecan (ACAN), pro-collagen-I alpha (Pro-Col-Iα), cartilage oligomeric matrix protein (COMP), interleukin-6 (IL-6) and vascular endothelial growth factor (VEGF) was assessed by enzyme-linked immunosorbent assay (ELISA). Labrum calcification was evaluated by 3D whole mount fluorescent microscopy of ethyl cinnamate-based optically cleared tissues stained with Alcian blue/Alizarin red. Results Whole mount microscopy revealed non-OA fracture controls were non-calcified, whereas six OA labra (40%) were partially calcified or ossified. Basal secretion of Pro-Col-Iα and VEGF was increased four-fold in OA versus non-OA labra. Pro-Col-Iα levels were correlated with those of VEGF (r = 0.65) and COMP (r = 0.54). Stimulation of DAMP signaling through TLR4 affected secretion of IL-6, VEGF, COMP and Pro-Col-Iα, with distinct responses between non-OA and OA tissues. Inhibition of TGF-beta signaling specifically reduced elevated secretion of Pro-Col- Iα and VEGF in calcified OA labrum. Conclusions Secretion of the putative OA biomarkers Pro-Col-Iα and VEGF is elevated in degenerated human acetabular labrum and may serve as indicators of OA and calcification status. Secretion of both factors was partially regulated by TGF-beta signaling in calcified OA labrum tissues.The Translational potential of this article:Our findings suggest that a biomarker panel consisting of Pro-Col-Iα/VEGF/COMP may be valuable for assessing subradiographic labrum degeneration and calcification in hip OA. Targeting TGF-beta signaling may offer a means to reduce vascular invasion and fibrosis in acetabular labrum tissue.
Collapse
Affiliation(s)
- Alexander Antoniadis
- Department of Orthopedics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Julien Wegrzyn
- Department of Orthopedics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Patrick Omoumi
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Léa Loisay
- Department of Rheumatology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Thomas Hügle
- Department of Rheumatology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Jeroen Geurts
- Department of Rheumatology, Lausanne University Hospital and University of Lausanne, Switzerland
| |
Collapse
|
5
|
Zhang X, Wang Z, Wang B, Li J, Yuan H. lncRNA OIP5-AS1 attenuates the osteoarthritis progression in IL-1β-stimulated chondrocytes. Open Med (Wars) 2023; 18:20230721. [PMID: 37333451 PMCID: PMC10276615 DOI: 10.1515/med-2023-0721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 04/18/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
In view of the association between long noncoding RNA OIP5-AS1 and osteoarthritis (OA) pathology, the corresponding potential mechanism is worthy of exploration. Primary chondrocytes were identified by morphological observation and immunohistochemical staining of collagen II. The association between OIP5-AS1 and miR-338-3p was analyzed by StarBase and dual-luciferase reporter assay. After the expression of OIP5-AS1 or miR-338-3p in interleukin (IL)-1β-stimulated primary chondrocytes and CHON-001 cells was manipulated, cell viability, proliferation, apoptosis rate, apoptosis-related protein (cleaved caspase-9, Bax) expressions, extracellular matrix (ECM) (matrix metalloproteinase (MMP)-3, MMP-13, aggrecan, and collagen II), PI3K/AKT pathway, and mRNA expressions of inflammatory factors (IL-6 and IL-8), OIP5-AS1, and miR-338-3p were determined by cell counting kit-8, EdU, flow cytometry, Western blot, and quantitative reverse transcription-polymerase chain reaction. As a result, the expression of OIP5-AS1 was downregulated in IL-1β-activated chondrocytes, while miR-338-3p was overexpressed. OIP5-AS1 overexpression reversed the effects of IL-1β on viability, proliferation, apoptosis, ECM degradation, and inflammation in chondrocytes. However, OIP5-AS1 knockdown exhibited opposite effects. Interestingly, the effects of OIP5-AS1 overexpression were partially offset by miR-338-3p overexpression. Furthermore, OIP5-AS1 overexpression blocked the PI3K/AKT pathway by modulating miR-338-3p expression. In sum, OIP5-AS1 promotes viability and proliferation, and inhibits apoptosis and ECM degradation in IL-1β-activated chondrocytes by targeting miR-338-3p through blocking the PI3K/AKT pathway, indicating an attractive strategy for OA treatment.
Collapse
Affiliation(s)
- Xuefeng Zhang
- The First Clinical Medical College, Jinan University, Guangzhou, Guangdong 510632, P.R. China
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Zhikun Wang
- Department of Orthopedics, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Binbin Wang
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Jingyi Li
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| | - Hui Yuan
- Department of Pain, SSL Central Hospital of Dongguan, Affiliated Dongguan Shilong People’s Hospital of Southern Medical University, Dongguan, Guangdong 523326, P.R. China
| |
Collapse
|
6
|
Rydén M, Önnerfjord P. In Vitro Models and Proteomics in Osteoarthritis Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1402:57-68. [PMID: 37052846 DOI: 10.1007/978-3-031-25588-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
This review summarizes and exemplifies the current understanding of osteoarthritis in vitro models and describes their relevance for new insights in the future of osteoarthritis research. Our friend and highly appreciated colleague, Prof. Alan Grodzinsky has contributed greatly to the understanding of joint tissue biology and cartilage biomechanics. He frequently utilizes in vitro models and cartilage explant cultures, and recent work also includes proteomics studies. This review is dedicated to honor his 75-year birthday and will focus on recent proteomic in vitro studies related to osteoarthritis, and within this topic highlight some of his contributions to the field.
Collapse
Affiliation(s)
- Martin Rydén
- Orthopaedics, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
7
|
Dou H, Wang S, Hu J, Song J, Zhang C, Wang J, Xiao L. Osteoarthritis models: From animals to tissue engineering. J Tissue Eng 2023; 14:20417314231172584. [PMID: 37223125 PMCID: PMC10201005 DOI: 10.1177/20417314231172584] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/13/2023] [Indexed: 05/25/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative osteoarthropathy. Although it has been revealed that a variety of factors can cause or aggravate the symptoms of OA, the pathogenic mechanisms of OA remain unknown. Reliable OA models that accurately reflect human OA disease are crucial for studies on the pathogenic mechanism of OA and therapeutic drug evaluation. This review first demonstrated the importance of OA models by briefly introducing the OA pathological features and the current limitations in the pathogenesis and treatment of OA. Then, it mainly discusses the development of different OA models, including animal and engineered models, highlighting their advantages and disadvantages from the perspective of pathogenesis and pathology analysis. In particular, the state-of-the-art engineered models and their potential were emphasized, as they may represent the future direction in the development of OA models. Finally, the challenges in obtaining reliable OA models are also discussed, and possible future directions are outlined to shed some light on this area.
Collapse
Affiliation(s)
- Hongyuan Dou
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Shuhan Wang
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical Devices, Shenzhen, China
| | - Jiawei Hu
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Jian Song
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
8
|
Yang D, Cao G, Ba X, Jiang H. Epigallocatechin-3- O-gallate promotes extracellular matrix and inhibits inflammation in IL-1β stimulated chondrocytes by the PTEN/miRNA-29b pathway. PHARMACEUTICAL BIOLOGY 2022; 60:589-599. [PMID: 35260041 PMCID: PMC8920401 DOI: 10.1080/13880209.2022.2039722] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
CONTEXT Epigallocatechin-3-O-gallate (EGCG) exhibits anti-arthritic activity. MiR-29b-3p provokes chondrocyte apoptosis and promotes the initiation and development of osteoarthritis (OA). OBJECTIVE To explore the roles of EGCG and miR-29b-3p in interleukin-1β (IL-1β)-stimulated chondrocytes. MATERIALS AND METHODS HE and Safranin O staining were used to detect the pathological changes of cartilage tissue in OA patients and healthy people. OA-like chondrocyte injury was mimicked by 5 ng/mL IL-1β stimulation for 24 h in vitro, and after transfection with miR-29b-3p mimics and pcDNA-PTEN, IL-1β-stimulated chondrocytes were pre-treated with EGCG (20 and 50 μM) for 2 h. Cell viability, colony numbers, apoptosis rate, the levels of IL-6 and matrix metalloproteinase-13 (MMP-13), miR-19b-3p, PTEN and apoptosis-associated proteins in chondrocytes were evaluated. RESULTS MiR-29b-3p level was upregulated in cartilage tissues of OA patients (3.5-fold change, p < 0.001) and IL-1β stimulated chondrocytes (two fold change, p < 0.001). The matrix staining was weakened and unevenly distributed, and the chondrocytes were arranged disorderly in the tissues of patients with OA. EGCG (20 and 50 μM) increases viability and decreases the levels of miR-29b-3p and MMP-13 and IL-6 in IL-1β stimulated chondrocytes (p < 0.05). MiR-29b-3p mimics reversed the effects above 50 μM EGCG (p < 0.05). Furthermore, PTEN overexpression abrogated the effects of miR-29b-3p mimics on viability, colony numbers, apoptosis rate and the levels of Bcl-2, MMP-13, IL-6, Bax and cleaved caspase 3 in IL-1β-stimulated chondrocytes (p < 0.01). DISCUSSION AND CONCLUSIONS EGCG is a potential candidate for the treatment of OA, which also can be explored in a novel therapeutic method for other degenerative or inflammatory disorders.
Collapse
Affiliation(s)
- Dong Yang
- Department of Orthopaedics, Liyang Peoples’ Hospital, Changzhou, PR China
- CONTACT Dong Yang Department of Orthopaedics, Liyang Peoples’ Hospital, No. 70, Jianshe West Road, Liyang, Changzhou, Jiangsu Province213361, PR China
| | - Guanghua Cao
- Department of Orthopaedics, Liyang Peoples’ Hospital, Changzhou, PR China
| | - Xiaorong Ba
- Department of Orthopaedics, Liyang Peoples’ Hospital, Changzhou, PR China
| | - Haibo Jiang
- Department of Orthopaedics, Liyang Peoples’ Hospital, Changzhou, PR China
| |
Collapse
|
9
|
Black RM, Flaman LL, Lindblom K, Chubinskaya S, Grodzinsky AJ, Önnerfjord P. Tissue catabolism and donor-specific dexamethasone response in a human osteochondral model of post-traumatic osteoarthritis. Arthritis Res Ther 2022; 24:137. [PMID: 35689293 PMCID: PMC9185927 DOI: 10.1186/s13075-022-02828-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/22/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Post-traumatic osteoarthritis (PTOA) does not currently have clinical prognostic biomarkers or disease-modifying drugs, though promising candidates such as dexamethasone (Dex) exist. Many challenges in studying and treating this disease stem from tissue interactions that complicate understanding of drug effects. We present an ex vivo human osteochondral model of PTOA to investigate disease effects on cartilage and bone homeostasis and discover biomarkers for disease progression and drug efficacy. METHODS Human osteochondral explants were harvested from normal (Collins grade 0-1) ankle talocrural joints of human donors (2 female, 5 male, ages 23-70). After pre-equilibration, osteochondral explants were treated with a single-impact mechanical injury and TNF-α, IL-6, and sIL-6R ± 100 nM Dex for 21 days and media collected every 2-3 days. Chondrocyte viability, tissue DNA content, and glycosaminoglycan (sGAG) percent loss to the media were assayed and compared to untreated controls using a linear mixed effects model. Mass spectrometry analysis was performed for both cartilage tissue and pooled culture medium, and the statistical significance of protein abundance changes was determined with the R package limma and empirical Bayes statistics. Partial least squares regression analyses of sGAG loss and Dex attenuation of sGAG loss against proteomic data were performed. RESULTS Injury and cytokine treatment caused an increase in the release of matrix components, proteases, pro-inflammatory factors, and intracellular proteins, while tissue lost intracellular metabolic proteins, which was mitigated with the addition of Dex. Dex maintained chondrocyte viability and reduced sGAG loss caused by injury and cytokine treatment by 2/3 overall, with donor-specific differences in the sGAG attenuation effect. Biomarkers of bone metabolism had mixed effects, and collagen II synthesis was suppressed with both disease and Dex treatment by 2- to 5-fold. Semitryptic peptides associated with increased sGAG loss were identified. Pro-inflammatory humoral proteins and apolipoproteins were associated with lower Dex responses. CONCLUSIONS Catabolic effects on cartilage tissue caused by injury and cytokine treatment were reduced with the addition of Dex in this osteochondral PTOA model. This study presents potential peptide biomarkers of early PTOA progression and Dex efficacy that can help identify and treat patients at risk of PTOA.
Collapse
Affiliation(s)
- Rebecca Mae Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Lisa L Flaman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Karin Lindblom
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Susan Chubinskaya
- Departments of Pediatrics, Orthopedic Surgery and Medicine (Section of Rheumatology), Rush University Medical Center, Chicago, IL, USA
| | - Alan J Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Patrik Önnerfjord
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
10
|
Pan Y, Yang Y, Fan M, Chen C, Jiang R, Liang L, Xian M, Kuang B, Geng N, Feng N, Deng L, Zheng W, Zhang F, Li X, Guo F. Progranulin regulation of autophagy contributes to its chondroprotective effect in osteoarthritis. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
11
|
Seidel MF, Netzer C, Chobaz V, Hügle T, Geurts J. Localization of Nerve Growth Factor Expression to Structurally Damaged Cartilaginous Tissues in Human Lumbar Facet Joint Osteoarthritis. Front Immunol 2022; 13:783076. [PMID: 35300334 PMCID: PMC8921992 DOI: 10.3389/fimmu.2022.783076] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 02/04/2022] [Indexed: 01/25/2023] Open
Abstract
Purpose Nerve Growth Factor (NGF) is a pivotal mediator of chronic pain and plays a role in bone remodelling. Through its high affinity receptor TrkA, NGF induces substance P (SP) as key downstream mediator of pain and local inflammation. Here we analysed NGF, TrkA and SP tissue distribution in facet joint osteoarthritis (FJOA), a major cause of chronic low back pain. Methods FJOA specimens (n=19) were harvested from patients undergoing intervertebral fusion surgery. Radiologic grading of FJOA and spinal stenosis, followed by immunohistochemistry for NGF, TrkA and SP on consecutive tissue sections, was performed in ten specimens. Explant cultures (n=9) were used to assess secretion of NGF, IL-6, and SP by FJOA osteochondral tissues under basal and inflammatory conditions. Results NGF was predominantly expressed in damaged cartilaginous tissues (80%), occasionally in bone marrow (20%), but not in osteochondral vascular channels. NGF area fraction in cartilage was not associated with the extent of proteoglycan loss or radiologic FJOA severity. Consecutive sections showed that NGF and SP expression was localized at structurally damaged cartilage, in absence of TrkA expression. SP and TrkA were expressed in subchondral bone marrow in both presence and absence of NGF. Low level NGF, but not SP secretion, was detected in four out of eighteen FJOA explants under both basal or inflammatory conditions (n=2 each). Conclusion NGF is associated with SP expression and structural cartilage damage in osteoarthritic facet joints, but not with radiologic disease severity. NGF tissue distribution in FJOA differs from predominant subchondral bone expression reported for knee OA.
Collapse
Affiliation(s)
- Matthias F Seidel
- Department of Rheumatology, Spitalzentrum-Centre Hospitalier, Biel-Bienne, Switzerland
| | - Cordula Netzer
- Spine Surgery, Department of Biomedical Engineering, University Hospital of Basel, Basel, Switzerland
| | - Véronique Chobaz
- Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Thomas Hügle
- Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Jeroen Geurts
- Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
12
|
Xiong Y, He Y, Peng Y, Geng Y. Association of IL-6 and TGF-β Gene Polymorphisms with the Risk of Thoracolumbar Osteoporotic Vertebral Compression Fractures. Pharmgenomics Pers Med 2022; 15:351-358. [PMID: 35469148 PMCID: PMC9034889 DOI: 10.2147/pgpm.s351372] [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: 11/26/2021] [Accepted: 02/09/2022] [Indexed: 12/01/2022] Open
Abstract
Purpose Osteoporotic vertebral compression fracture (OVCF) is a common disease in the elderly, and genetic factors play a key role in its occurrence. The present study was conducted to investigate the association between interleukin-6 (IL-6) and the transforming growth factor (TGF-β) gene polymorphisms and the occurrence of thoracolumbar OVCF. Patients and Methods This case–control study recruited 146 patients with OVCF and 144 osteoporosis patients as the control group. Genotypes of the IL-6 rs1800796 and TGF-β rs1982073 were analyzed by sequencing. Genotype distribution and allelic frequencies were investigated by the χ2 test. Odds ratios (OR) and 95% confidence intervals (CI) evaluated the relationship of IL-6 or TGF-β polymorphism and OVCF susceptibility. Results Allele G and genotype GG of IL-6 rs1800796 was more frequent in patients with OVCF (40.07% vs.28.47%; 19.18% vs.7.64%) compared with controls. GG genotype (OR=3.394, 95% CI=1.560–7.385, P < 0.001) and G allele (OR=1.680, 95% CI=1.187–2.376, P < 0.001) of IL-6 rs1800796 was significantly associated with increased risk of OVCF. What is more, CT and TT genotypes (41.78 vs.51.39; 19.86 vs.26.39) and allele T (40.75 vs 52.08) of TGF-β rs1982073 were less frequent in OVCFs, more common in controls and protective against OVCF risk (OR=0.436, 95% CI=0.228–0.835, P = 0.012; OR=0.615, 95% CI=0.443–0.855, P = 0.004). Conclusion Our results suggest that the G allele and GG genotype of IL-6 rs1800796 may contribute to increased susceptibility to OVCF in elderly Chinese. In contrast, CT and TT genotypes and the T allele of TGF-β rs1982073 may contribute to lower susceptibility of OVCF.
Collapse
Affiliation(s)
- Yi Xiong
- Department of Orthopaedic, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Hubei, People’s Republic of China
| | - Ye He
- Department of Preventive Treatment of Diseases,Shaanxi Meixian Hospital of Traditional Chinese Medicine, Shaanxi, People’s Republic of China
| | - Yan Peng
- Department of Medical Examination, Yili Kazak Autonomous Prefecture Hospital of Traditional Chinese Medicine, Xinjiang, People’s Republic of China
| | - Yun Geng
- Department of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong, People’s Republic of China
- Correspondence: Yun Geng, Tel/Fax +86-531-59556066, Email
| |
Collapse
|
13
|
Muenzebrock KA, Kersten V, Alblas J, Garcia JP, Creemers LB. The Added Value of the “Co” in Co-Culture Systems in Research on Osteoarthritis Pathology and Treatment Development. Front Bioeng Biotechnol 2022; 10:843056. [PMID: 35309991 PMCID: PMC8927651 DOI: 10.3389/fbioe.2022.843056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent disease and a major health burden. Its development and progression are influenced by factors such as age, obesity or joint overuse. As a whole organ disease OA affects not only cartilage, bone and synovium but also ligaments, fatty or nervous tissue surrounding the joint. These joint tissues interact with each other and understanding this interaction is important in developing novel treatments. To incorporate and study these interactions in OA research, several co-culture models have evolved. They combine two or more cell types or tissues and investigate the influence of amongst others inflammatory or degenerative stimuli seen in OA. This review focuses on co-cultures and the differential processes occurring in a given tissue or cell as a consequence of being combined with another joint cell type or tissue, and/or the extent to which a co-culture mimics the in vivo processes. Most co-culture models depart from synovial lining and cartilage culture, but also fat pad and bone have been included. Not all of the models appear to reflect the postulated in vivo OA pathophysiology, although some of the discrepancies may indicate current assumptions on this process are not entirely valid. Systematic analysis of the mutual influence the separate compartments in a given model exert on each other and validation against in vivo or ex vivo observation is still largely lacking and would increase their added value as in vitro OA models.
Collapse
|
14
|
Papaneophytou C, Alabajos-Cea A, Viosca-Herrero E, Calvis C, Costa M, Christodoulides AE, Kroushovski A, Lapithis A, Lapithi VM, Papayiannis I, Christou A, Messeguer R, Giannaki C, Felekkis K. Associations between serum biomarkers of cartilage metabolism and serum hyaluronic acid, with risk factors, pain categories, and disease severity in knee osteoarthritis: a pilot study. BMC Musculoskelet Disord 2022; 23:195. [PMID: 35236298 PMCID: PMC8889762 DOI: 10.1186/s12891-022-05133-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Specific serum biomarkers of cartilage metabolism such as cartilage oligomeric matrix protein (sCOMP) and procollagen type II C-terminal propeptide (sPIICP) as well as hyaluronan (sHA), a biomarker of synovitis, have been implicated in the pathophysiology of knee osteoarthritis (OA). However, the associations of these biomarkers with the severity of the disease and OA risk factors, including age and obesity remain inconclusive. This analysis examines the associations between these serum biomarkers and the radiographic severity of OA and knee pain, as wells as obesity, the age and gender of the participants, and other OA risk factors. METHODS From 44 patients with early knee OA and 130 patients with late knee OA we analyzed the radiographic severity of the disease using the Kellgren and Lawrence (KL) grading system. Moreover, 38 overweight healthy individuals were used as a control group. Specific information was collected from all participants during their recruitment. The levels of the three serum biomarkers were quantified using commercially available ELISA kits. Serum biomarkers were analyzed for associations with the average KL scores and pain in both knees, as well as with specific OA risk factors. RESULTS The levels of sCOMP were elevated in patients with severe late OA and knee pain and correlated weakly with OA severity. A weakly correlation of sHA levels and OA severity OA was observed. We demonstrated that only sPIICP levels were markedly decreased in patients with late knee OA suggesting the alterations of cartilage metabolism in this arthritic disease. Moreover, we found that sPIICP has the strongest correlation with obesity and the severity of OA, as well as with the knee pain at rest and during walking regardless of the severity of the disease. ROC analysis showed that the area under the ROC curve (AUC) was 0.980 (95% CI: 0.945-0.995; p < 0.0001), suggesting high diagnostic accuracy of sPIICP. Interestingly, gender and age had also an effect on the levels of sPIICP. CONCLUSION This study revealed the potential of serum PIICP to be used as a biomarker to monitor the progression of knee OA, however, further studies are warranted to elucidate its clinical implication.
Collapse
Affiliation(s)
- Christos Papaneophytou
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417, Nicosia, Cyprus
| | - Ana Alabajos-Cea
- Physical Medicine & Rehabilitation Department, Hospital La Fe, 46026, Valencia, Spain
- Health Research Institute La Fe, 46026, Valencia, Spain
| | | | - Carme Calvis
- Drug Development Area, Health & Biomedicine Department, LEITAT Technological Centre, Parc Científic de Barcelona, 08028, Barcelona, Spain
| | - Marta Costa
- Drug Development Area, Health & Biomedicine Department, LEITAT Technological Centre, Parc Científic de Barcelona, 08028, Barcelona, Spain
| | | | - Alexander Kroushovski
- Apollonion Hospital, 2054, Nicosia, Cyprus
- Medical School, University of Nicosia, 2408, Nicosia, Cyprus
| | | | | | | | | | - Ramon Messeguer
- Drug Development Area, Health & Biomedicine Department, LEITAT Technological Centre, Parc Científic de Barcelona, 08028, Barcelona, Spain
| | - Christoforos Giannaki
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417, Nicosia, Cyprus.
| | - Kyriacos Felekkis
- Department of Life and Health Sciences, School of Sciences and Engineering, University of Nicosia, 2417, Nicosia, Cyprus.
| |
Collapse
|
15
|
Li K, Zhang P, Zhu Y, Alini M, Grad S, Li Z. Establishment of an Ex Vivo Inflammatory Osteoarthritis Model With Human Osteochondral Explants. Front Bioeng Biotechnol 2022; 9:787020. [PMID: 34993189 PMCID: PMC8724558 DOI: 10.3389/fbioe.2021.787020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/25/2021] [Indexed: 01/16/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease without clear pathophysiological mechanism and effective drugs for treatment. Although various animal models exist, the translation of the outcome into clinics remains difficult due to species differences. In this study, an ex vivo inflammatory OA model was induced using different concentrations of interleukin one beta (IL-1β) and tumor necrosis factor α (TNF-α) on explants from the human femoral head. In the inflammatory OA groups, the gene expression levels of cartilage catabolism (matrix metalloproteinase 1 (MMP1), matrix metalloproteinase 3 (MMP3)), and inflammation (interleukin 6 (IL-6), interleukin 8 (IL-8)) markers were significantly upregulated, while the anabolic genes (collagen 2 (COL2), aggrecan (ACAN), and proteoglycan 4 (PRG4)) were downregulated compared to the control group. The release of cytokines (IL-6, IL-8) and nitric oxide (NO) in the conditioned medium was also upregulated in inflammatory OA groups. The Safranin O/Fast Green staining showed loss of proteoglycan in the superficial zone cartilage after cytokine treatment. The results indicated that an ex vivo inflammation and degeneration model was successfully established using osteochondral explants from the human femoral head. This model can be used to elucidate the in-depth mechanism of inflammatory OA and to screen new drugs for OA treatment.
Collapse
Affiliation(s)
- Kaihu Li
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China.,AO Research Institute Davos, Davos, Switzerland
| | - Penghui Zhang
- AO Research Institute Davos, Davos, Switzerland.,Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yong Zhu
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | - Zhen Li
- AO Research Institute Davos, Davos, Switzerland
| |
Collapse
|
16
|
Styczynska-Soczka K, Amin AK, Simpson AHW, Hall AC. Optimization and Validation of a Human Ex Vivo Femoral Head Model for Preclinical Cartilage Research and Regenerative Therapies. Cartilage 2021; 13:386S-397S. [PMID: 32567330 PMCID: PMC8721618 DOI: 10.1177/1947603520934534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Articular cartilage is incapable of effective repair following injury or during osteoarthritis. While there have been developments in cartilage repair technologies, there is a need to advance biologically relevant models for preclinical testing of biomaterial and regenerative therapies. This study describes conditions for the effective ex vivo culture of the whole human femoral head. DESIGN Fresh, viable femoral heads were obtained from femoral neck fractures and cultured for up to 10 weeks in (a) Dulbecco's modified Eagle's medium (DMEM); (b) DMEM + mixing; (c) DMEM + 10% human serum (HS); (d) DMEM + 10% HS + mixing. The viability, morphology, volume, and density of fluorescently labelled in situ chondrocytes and cartilage surface roughness were assessed by confocal microscopy. Cartilage histology was studied for glycosaminoglycan content using Alcian blue and collagen content using picrosirius red. RESULTS Chondrocyte viability remained at >95% in DMEM + 10% HS. In DMEM alone, viability remained high for ~4 weeks and then declined. For the other conditions, superficial zone chondrocyte viability fell to <35% at 10 weeks with deeper zones being relatively unaffected. In DMEM + 10% HS at 10 weeks, the number of chondrocytes possessing cytoplasmic processes increased compared with DMEM (P = 0.017). Alcian blue labeling decreased (P = 0.02) and cartilage thinned (P ≤ 0.05); however, there was no change to surface roughness, chondrocyte density, chondrocyte volume, or picrosirius red labeling (P > 0.05). CONCLUSIONS In this ex vivo model, chondrocyte viability was maintained in human femoral heads for up to 10 weeks in culture, a novel finding not previously reported. This human model could prove invaluable for the exploration, development, and assessment of preclinical cartilage repair and regenerative therapies.
Collapse
Affiliation(s)
| | - Anish K. Amin
- Department of Trauma and Orthopaedic
Surgery, Royal Infirmary of Edinburgh, Edinburgh, Scotland, UK
| | - A. Hamish W. Simpson
- Department of Trauma and Orthopaedic
Surgery, Royal Infirmary of Edinburgh, Edinburgh, Scotland, UK
| | - Andrew C. Hall
- Biomedical Sciences, Edinburgh Medical
School, University of Edinburgh, Edinburgh, Scotland, UK,Andrew C. Hall, Biomedical Sciences,
Edinburgh Medical School, University of Edinburgh, Hugh Robson Building, George
Square, Edinburgh, EH8 9XD, Scotland, UK.
| |
Collapse
|
17
|
Singh YP, Moses JC, Bhardwaj N, Mandal BB. Overcoming the Dependence on Animal Models for Osteoarthritis Therapeutics - The Promises and Prospects of In Vitro Models. Adv Healthc Mater 2021; 10:e2100961. [PMID: 34302436 DOI: 10.1002/adhm.202100961] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/10/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis (OA) is a musculoskeletal disease characterized by progressive degeneration of osteochondral tissues. Current treatment is restricted to the reduction of pain and loss of function of the joint. To better comprehend the OA pathophysiological conditions, several models are employed, however; there is no consensus on a suitable model. In this review, different in vitro models being developed for possible therapeutic intervention of OA are outlined. Herein, various in vitro OA models starting from 2D model, co-culture model, 3D models, dynamic culture model to advanced technologies-based models such as 3D bioprinting, bioassembly, organoids, and organ-on-chip-based models are discussed with their advantages and disadvantages. Besides, different growth factors, cytokines, and chemicals being utilized for induction of OA condition are reviewed in detail. Furthermore, there is focus on scrutinizing different molecular and possible therapeutic targets for better understanding the mechanisms and OA therapeutics. Finally, the underlying challenges associated with in vitro models are discussed followed by future prospective. Taken together, a comprehensive overview of in vitro OA models, factors to induce OA-like conditions, and intricate molecular targets with the potential to develop personalized osteoarthritis therapeutics in the future with clinical translation is provided.
Collapse
Affiliation(s)
- Yogendra Pratap Singh
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Joseph Christakiran Moses
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Nandana Bhardwaj
- Department of Science and Mathematics Indian Institute of Information Technology Guwahati Bongora Guwahati Assam 781015 India
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Sciences and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
18
|
Lv Z, Yang YX, Li J, Fei Y, Guo H, Sun Z, Lu J, Xu X, Jiang Q, Ikegawa S, Shi D. Molecular Classification of Knee Osteoarthritis. Front Cell Dev Biol 2021; 9:725568. [PMID: 34513847 PMCID: PMC8429960 DOI: 10.3389/fcell.2021.725568] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/09/2021] [Indexed: 01/15/2023] Open
Abstract
Knee osteoarthritis (KOA) is the most common form of joint degeneration with increasing prevalence and incidence in recent decades. KOA is a molecular disorder characterized by the interplay of numerous molecules, a considerable number of which can be detected in body fluids, including synovial fluid, urine, and blood. However, the current diagnosis and treatment of KOA mainly rely on clinical and imaging manifestations, neglecting its molecular pathophysiology. The mismatch between participants' molecular characteristics and drug therapeutic mechanisms might explain the failure of some disease-modifying drugs in clinical trials. Hence, according to the temporal alteration of representative molecules, we propose a novel molecular classification of KOA divided into pre-KOA, early KOA, progressive KOA, and end-stage KOA. Then, progressive KOA is furtherly divided into four subtypes as cartilage degradation-driven, bone remodeling-driven, inflammation-driven, and pain-driven subtype, based on the major pathophysiology in patient clusters. Multiple clinical findings of representatively investigated molecules in recent years will be reviewed and categorized. This molecular classification allows for the prediction of high-risk KOA individuals, the diagnosis of early KOA patients, the assessment of therapeutic efficacy, and in particular, the selection of homogenous patients who may benefit most from the appropriate therapeutic agents.
Collapse
Affiliation(s)
- Zhongyang Lv
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yannick Xiaofan Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiawei Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yuxiang Fei
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hu Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ziying Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jun Lu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xingquan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Science (IMS, RIKEN), Tokyo, Japan
| | - Dongquan Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
19
|
Samvelyan HJ, Hughes D, Stevens C, Staines KA. Models of Osteoarthritis: Relevance and New Insights. Calcif Tissue Int 2021; 109:243-256. [PMID: 32062692 PMCID: PMC8403120 DOI: 10.1007/s00223-020-00670-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/05/2020] [Indexed: 12/31/2022]
Abstract
Osteoarthritis (OA) is a progressive and disabling musculoskeletal disease affecting millions of people and resulting in major healthcare costs worldwide. It is the most common form of arthritis, characterised by degradation of the articular cartilage, formation of osteophytes, subchondral sclerosis, synovial inflammation and ultimate loss of joint function. Understanding the pathogenesis of OA and its multifactorial aetiology will lead to the development of effective treatments, which are currently lacking. Two-dimensional (2D) in vitro tissue models of OA allow affordable, high-throughput analysis and stringent control over specific variables. However, they are linear in fashion and are not representative of physiological conditions. Recent in vitro studies have adopted three-dimensional (3D) tissue models of OA, which retain the advantages of 2D models and are able to mimic physiological conditions, thereby allowing investigation of additional variables including interactions between the cells and their surrounding extracellular matrix. Numerous spontaneous and induced animal models are used to reproduce the onset and monitor the progression of OA based on the aetiology under investigation. This therefore allows elucidation of the pathogenesis of OA and will ultimately enable the development of novel and specific therapeutic interventions. This review summarises the current understanding of in vitro and in vivo OA models in the context of disease pathophysiology, classification and relevance, thus providing new insights and directions for OA research.
Collapse
Affiliation(s)
| | - David Hughes
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh, UK
| | - Craig Stevens
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh, UK
| | - Katherine Ann Staines
- School of Applied Sciences, Edinburgh Napier University, Sighthill Campus, Edinburgh, UK.
| |
Collapse
|
20
|
Yamanashi Y, Ohmichi M, Ohmichi Y, Ikemoto T, Arai YC, Maruyama Y, Otsuka S, Hirai S, Naito M, Deie M. Efficacy of Methotrexate on Rat Knee Osteoarthritis Induced by Monosodium Iodoacetate. J Inflamm Res 2021; 14:3247-3259. [PMID: 34290513 PMCID: PMC8289442 DOI: 10.2147/jir.s318540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/25/2021] [Indexed: 12/25/2022] Open
Abstract
Objective To explore whether methotrexate (MTX) prevents joint destruction and improves pain-related behaviors in the acute phase of knee osteoarthritis (OA) induced by monosodium iodoacetate (MIA) in a rat model. Methods Twenty of 25 male Wistar rats (10–14 weeks old) received 3 mg MIA via intra-articular injection into their right knee and were then administered a vehicle control (n=10) or 3 mg/kg MTX orally weekly (n=10). We assessed differences in pain-related behavior, spontaneous lifting behavior, micro-computed tomography (CT), histopathology, and expression of pain- and inflammatory-related genes using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) between the two groups for 4 weeks. Five rats were used as untreated controls to assess pain- and inflammatory-related mRNA expression in the dorsal root ganglia (DRG) and knee joints using RT-qPCR. Results Joint destruction and mechanical hyperalgesia were observed in the vehicle group. Decreases in mechanical pain thresholds for the knee joint and calf muscles were improved after MTX administration; however, joint damage assessed by micro-CT and histopathology was not significantly inhibited by MTX administration, while upregulation levels of transient receptor potential cation channel, subfamily V, member 1 (TRPV-1) (P<0.01) and brain-derived neurotrophic factor (BDNF) (P=0.02) mRNA in the DRG and nerve growth factor NGF mRNA (P=0.03) in the affected knee joints were significantly suppressed in the MTX group compared with the vehicle group at week 4. Conclusion Our results imply that MTX administration improves pain-related behaviors and suppresses expression of pain-related mRNAs in the DRG and knee joint; however, MTX is not expected to prevent cartilage degeneration in MIA-induced OA in rat knee.
Collapse
Affiliation(s)
- Yuki Yamanashi
- Department of Orthopedic Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Mika Ohmichi
- Department of Anatomy II, Kanazawa Medical University, Kahoku, Ishikawa, Japan.,Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yusuke Ohmichi
- Department of Anatomy II, Kanazawa Medical University, Kahoku, Ishikawa, Japan.,Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Tatsunori Ikemoto
- Department of Orthopedic Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Young-Chang Arai
- Institute of Physical Fitness, Sports Medicine and Rehabilitation, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yohei Maruyama
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Shun Otsuka
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Shuichi Hirai
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Munekazu Naito
- Department of Anatomy, Aichi Medical University, Nagakute, Aichi, Japan
| | - Masataka Deie
- Department of Orthopedic Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
21
|
Proteomic Analysis of Synovial Fibroblasts and Articular Chondrocytes Co-Cultures Reveals Valuable VIP-Modulated Inflammatory and Degradative Proteins in Osteoarthritis. Int J Mol Sci 2021; 22:ijms22126441. [PMID: 34208590 PMCID: PMC8235106 DOI: 10.3390/ijms22126441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disorder causing a great disability and a reduction in the quality of life. In OA, articular chondrocytes (AC) and synovial fibroblasts (SF) release innate-derived immune mediators that initiate and perpetuate inflammation, inducing cartilage extracellular matrix (ECM) degradation. Given the lack of therapies for the treatment of OA, in this study, we explore biomarkers that enable the development of new therapeutical approaches. We analyze the set of secreted proteins in AC and SF co-cultures by stable isotope labeling with amino acids (SILAC). We describe, for the first time, 115 proteins detected in SF-AC co-cultures stimulated by fibronectin fragments (Fn-fs). We also study the role of the vasoactive intestinal peptide (VIP) in this secretome, providing new proteins involved in the main events of OA, confirmed by ELISA and multiplex analyses. VIP decreases proteins involved in the inflammatory process (CHI3L1, PTX3), complement activation (C1r, C3), and cartilage ECM degradation (DCN, CTSB and MMP2), key events in the initiation and progression of OA. Our results support the anti-inflammatory and anti-catabolic properties of VIP in rheumatic diseases and provide potential new targets for OA treatment.
Collapse
|
22
|
Houtman E, van Hoolwerff M, Lakenberg N, Suchiman EHD, van der Linden-van der Zwaag E, Nelissen RGHH, Ramos YFM, Meulenbelt I. Human Osteochondral Explants: Reliable Biomimetic Models to Investigate Disease Mechanisms and Develop Personalized Treatments for Osteoarthritis. Rheumatol Ther 2021; 8:499-515. [PMID: 33608843 PMCID: PMC7991015 DOI: 10.1007/s40744-021-00287-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/30/2021] [Indexed: 02/07/2023] Open
Abstract
Introduction Likely due to ignored heterogeneity in disease pathophysiology, osteoarthritis (OA) has become the most common disabling joint disease, without effective disease-modifying treatment causing a large social and economic burden. In this study we set out to explore responses of aged human osteochondral explants upon different OA-related perturbing triggers (inflammation, hypertrophy and mechanical stress) for future tailored biomimetic human models. Methods Human osteochondral explants were treated with IL-1β (10 ng/ml) or triiodothyronine (T3; 10 nM) or received 65% strains of mechanical stress (65% MS). Changes in chondrocyte signalling were determined by expression levels of nine genes involved in catabolism, anabolism and hypertrophy. Breakdown of cartilage was measured by sulphated glycosaminoglycans (sGAGs) release, scoring histological changes (Mankin score) and mechanical properties of cartilage. Results All three perturbations (IL-1β, T3 and 65% MS) resulted in upregulation of the catabolic genes MMP13 and EPAS1. IL-1β abolished COL2A1 and ACAN gene expression and increased cartilage degeneration, reflected by increased Mankin scores and sGAGs released. Treatment with T3 resulted in a high and significant upregulation of the hypertrophic markers COL1A1, COL10A1 and ALPL. However, 65% MS increased sGAG release and detrimentally altered mechanical properties of cartilage. Conclusion We present consistent and specific output on three different triggers of OA. Perturbation with the pro-inflammatory IL-1β mainly induced catabolic chondrocyte signalling and cartilage breakdown, while T3 initiated expression of hypertrophic and mineralization markers. Mechanical stress at a strain of 65% induced catabolic chondrocyte signalling and changed cartilage matrix integrity. The major strength of our ex vivo models was that they considered aged, preserved, human cartilage of a heterogeneous OA patient population. As a result, the explants may reflect a reliable biomimetic model prone to OA onset allowing for development of different treatment modalities. Supplementary Information The online version contains supplementary material available at 10.1007/s40744-021-00287-y.
Collapse
Affiliation(s)
- Evelyn Houtman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Marcella van Hoolwerff
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Nico Lakenberg
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Eka H D Suchiman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Rob G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Yolande F M Ramos
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid Meulenbelt
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Novel therapies for damaged and diseased bone are being developed in a preclinical testing process consisting of in vitro cell experiments followed by in vivo animal studies. The in vitro results are often not representative of the results observed in vivo. This could be caused by the complexity of the natural bone environment that is missing in vitro. Ex vivo bone explant cultures provide a model in which cells are preserved in their native three-dimensional environment. Herein, it is aimed to review the current status of bone explant culture models in relation to their potential in complementing the preclinical evaluation process with specific attention paid to the incorporation of mechanical loading within ex vivo culture systems. RECENT FINDINGS Bone explant cultures are often performed with physiologically less relevant bone, immature bone, and explants derived from rodents, which complicates translatability into clinical practice. Mature bone explants encounter difficulties with maintaining viability, especially in static culture. The integration of mechanical stimuli was able to extend the lifespan of explants and to induce new bone formation. Bone explant cultures provide unique platforms for bone research and mechanical loading was demonstrated to be an important component in achieving osteogenesis ex vivo. However, more research is needed to establish a representative, reliable, and reproducible bone explant culture system that includes both components of bone remodeling, i.e., formation and resorption, in order to bridge the gap between in vitro and in vivo research in preclinical testing.
Collapse
Affiliation(s)
- E E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - K Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - S Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
24
|
Makarczyk MJ, Gao Q, He Y, Li Z, Gold MS, Hochberg MC, Bunnell BA, Tuan RS, Goodman SB, Lin H. Current Models for Development of Disease-Modifying Osteoarthritis Drugs. Tissue Eng Part C Methods 2021; 27:124-138. [PMID: 33403944 PMCID: PMC8098772 DOI: 10.1089/ten.tec.2020.0309] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a painful and disabling disease that affects millions of people worldwide. Symptom-alleviating treatments exist, although none with long-term efficacy. Furthermore, there are currently no disease-modifying OA drugs (DMOADs) with demonstrated efficacy in OA patients, which is, in part, attributed to a lack of full understanding of the pathogenesis of OA. The inability to translate findings from basic research to clinical applications also highlights the deficiencies in the available OA models at simulating the clinically relevant pathologies and responses to treatments in humans. In this review, the current status in the development of DMOADs will be first presented, with special attention to those in Phase II-IV clinical trials. Next, current in vitro, ex vivo, and in vivo OA models are summarized and the respective advantages and disadvantages of each are highlighted. Of note, the development and application of microphysiological or tissue-on-a-chip systems for modeling OA in humans are presented and the issues that need to be addressed in the future are discussed. Microphysiological systems should be given serious consideration for their inclusion in the DMOAD development pipeline, both for their ability to predict drug safety and efficacy in human clinical trials at present, as well as for their potential to serve as a test platform for personalized medicine. Impact statement At present, no disease-modifying osteoarthritis (OA) drugs (DMOADs) have been approved for widespread clinical use by regulatory bodies. The failure of developing effective DMOADs is likely owing to multiple factors, not the least of which are the intrinsic differences between the intact human knee joint and the preclinical models. This work summarizes the current OA models for the development of DMOADs, discusses the advantages/disadvantages of each, and then proposes future model development to aid in the discovery of effective and personalized DMOADs. The review also highlights the microphysiological systems, which are emerging as a new platform for drug development.
Collapse
Affiliation(s)
- Meagan J. Makarczyk
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University, California, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhong Li
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael S. Gold
- Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark C. Hochberg
- Department of Medicine and Epidemiology and Public Health, University of Maryland, Baltimore, Maryland, USA
| | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Stuart B. Goodman
- Department of Orthopaedic Surgery, Stanford University, California, USA
- Department of Bioengineering, Stanford University, California, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
25
|
Salgado C, Jordan O, Allémann E. Osteoarthritis In Vitro Models: Applications and Implications in Development of Intra-Articular Drug Delivery Systems. Pharmaceutics 2021; 13:60. [PMID: 33466397 PMCID: PMC7824837 DOI: 10.3390/pharmaceutics13010060] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a complex multi-target disease with an unmet medical need for the development of therapies that slow and potentially revert disease progression. Intra-articular (IA) delivery has seen a surge in osteoarthritis research in recent years. As local administration of molecules, this represents a way to circumvent systemic drug delivery struggles. When developing intra-articular formulations, the main goals are a sustained and controlled release of therapeutic drug doses, taking into account carrier choice, drug molecule, and articular joint tissue target. Therefore, the selection of models is critical when developing local administration formulation in terms of accurate outcome assessment, target and off-target effects and relevant translation to in vivo. The current review highlights the applications of OA in vitro models in the development of IA formulation by means of exploring their advantages and disadvantages. In vitro models are essential in studies of OA molecular pathways, understanding drug and target interactions, assessing cytotoxicity of carriers and drug molecules, and predicting in vivo behaviors. However, further understanding of molecular and tissue-specific intricacies of cellular models for 2D and 3D needs improvement to accurately portray in vivo conditions.
Collapse
Affiliation(s)
- Carlota Salgado
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Olivier Jordan
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; (C.S.); (O.J.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
26
|
Zhang G, Zhang Q, Zhu J, Tang J, Nie M. LncRNA ARFRP1 knockdown inhibits LPS-induced the injury of chondrocytes by regulation of NF-κB pathway through modulating miR-15a-5p/TLR4 axis. Life Sci 2020; 261:118429. [PMID: 32931797 DOI: 10.1016/j.lfs.2020.118429] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022]
Abstract
AIMS Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have been reported as the important regulators in osteoarthritis (OA). However, the detailed mechanism is implicated. The aim of this study is to reveal the functional mechanism of lncRNA ARFRP1 and miR-15a-5p in osteoarthritis. MATERIALS AND METHODS The expression level of genes was detected by quantitative real time polymerase chain reaction (qRT-PCR) or western blot assay. Cell Counting Kit-8 (CCK-8) was used to assess cell viability. Cell apoptosis rate was analyzed by flow cytometry analysis. Furthermore, Enzyme-linked immunosorbent assay (ELISA) was performed to measure tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1β contents. The interaction between miR-15a-5p and ARFRP1 or Toll-like receptor 4 (TLR4) was predicted by miRcode or PITA, and then confirmed by the dual luciferase reporter assay or pull down assay. Besides, NF-κB-driven luciferase activity was determined using NF-κB luciferase reporter assay. KEY FINDINGS ARFRP1 and TLR4 levels were increased and miR-15a-5p level was decreased in OA cartilage tissues and lipopolysaccharides (LPS)-induced chondrocytes. ARFRP1 knockdown inhibited LPS-induced the injury of chondrocytes. Interestingly, miR-15a-5p downregulated by ARFRP1 negatively modulated TLR4 expression through interaction. ARFRP1 mediated LPS-induced the injury of chondrocytes via regulating miR-15a-5p/TLR4 axis. Furthermore, ARFRP1 exerted function by modulation of NF-κB pathway. SIGNIFICANCE Our findings confirmed that ARFRP1 mediated LPS-induced the injury of chondrocytes through regulating NF-κB pathway by modulation of miR-15a-5p/TLR4 axis, providing theoretical basis for the treatment of OA patients.
Collapse
Affiliation(s)
- Guangcheng Zhang
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Jiangsu, Zhejiang, China
| | - Qing Zhang
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Jiangsu, Zhejiang, China
| | - Juan Zhu
- Department of Anesthesiology, Affiliated Hospital of Jiangsu University, Jiangsu, Zhejiang, China
| | - Jiazhu Tang
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Jiangsu, Zhejiang, China
| | - Mingjun Nie
- Department of Orthopedics, Affiliated Hospital of Jiangsu University, Jiangsu, Zhejiang, China.
| |
Collapse
|
27
|
Profile of Matrix-Remodeling Proteinases in Osteoarthritis: Impact of Fibronectin. Cells 2019; 9:cells9010040. [PMID: 31877874 PMCID: PMC7017325 DOI: 10.3390/cells9010040] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a complex and specialized three-dimensional macromolecular network, present in nearly all tissues, that also interacts with cell surface receptors on joint resident cells. Changes in the composition and physical properties of the ECM lead to the development of many diseases, including osteoarthritis (OA). OA is a chronic degenerative rheumatic disease characterized by a progressive loss of synovial joint function as a consequence of the degradation of articular cartilage, also associated with alterations in the synovial membrane and subchondral bone. During OA, ECM-degrading enzymes, including urokinase-type plasminogen activator (uPA), matrix metalloproteinases (MMPs), and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs), cleave ECM components, such as fibronectin (Fn), generating fibronectin fragments (Fn-fs) with catabolic properties. In turn, Fn-fs promote activation of these proteinases, establishing a degradative and inflammatory feedback loop. Thus, the aim of this review is to update the contribution of ECM-degrading proteinases to the physiopathology of OA as well as their modulation by Fn-fs.
Collapse
|
28
|
Wu YR, Kuang GY, Lu FG, Wang HX, Lu M, Zhou Q. Pathological Relationship between Intestinal Flora and Osteoarthritis and Intervention Mechanism of Chinese Medicine. Chin J Integr Med 2019; 25:716-720. [PMID: 31650488 DOI: 10.1007/s11655-019-3224-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2019] [Indexed: 12/31/2022]
Abstract
Chinese medicine (CM) has a good clinical effect on osteoarthritis (OA), but the mechanism is not very clear. Evidence-based medicine researches have shown that intestinal flora plays a role in the pathogenesis and succession of OA. Intestinal flora affects the efficacy of CM, and CM can affect the balance of intestinal flora. This paper focuses on the relationship between intestinal flora, intestinal microenvironment, brain-gut axis, metabolic immunity and OA, and preliminarily expound the significance of intestinal flora in the pathogenesis of OA and the mechanism of CM intervention. The above discussion will be of great significance in the prevention and treatment of OA by CM from the perspective of intestinal flora.
Collapse
Affiliation(s)
- Yong-Rong Wu
- TAN Xin-hua Famous Medical Studio, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Gao-Yan Kuang
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Fang-Guo Lu
- Department of Medical College, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Heng-Xin Wang
- Hinye Pharmaceutical Co., Ltd., Changsha, 410331, China
| | - Min Lu
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Qing Zhou
- TAN Xin-hua Famous Medical Studio, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China.
| |
Collapse
|
29
|
Camacho-Encina M, Balboa-Barreiro V, Rego-Perez I, Picchi F, VanDuin J, Qiu J, Fuentes M, Oreiro N, LaBaer J, Ruiz-Romero C, Blanco FJ. Discovery of an autoantibody signature for the early diagnosis of knee osteoarthritis: data from the Osteoarthritis Initiative. Ann Rheum Dis 2019; 78:1699-1705. [PMID: 31471297 PMCID: PMC6900252 DOI: 10.1136/annrheumdis-2019-215325] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/23/2019] [Accepted: 08/22/2019] [Indexed: 11/27/2022]
Abstract
Objective To find autoantibodies (AAbs) in serum that could be useful to predict incidence of radiographic knee osteoarthritis (KOA). Design A Nucleic-acid Programmable Protein Arrays (NAPPA) platform was used to screen AAbs against 2125 human proteins in sera at baseline from participants free of radiographic KOA belonging to the incidence and non-exposed subcohorts of the Osteoarthritis Initiative (OAI) who developed or not, radiographic KOA during a follow-up period of 96 months. NAPPA-ELISA were performed to analyse reactivity against methionine adenosyltransferase two beta (MAT2β) and verify the results in 327 participants from the same subcohorts. The association of MAT2β-AAb levels with KOA incidence was assessed by combining several robust biostatistics analysis (logistic regression, Receiver Operating Characteristic and Kaplan-Meier curves). The proposed prognostic model was replicated in samples from the progression subcohort of the OAI. Results In the screening phase, six AAbs were found significantly different at baseline in samples from incident compared with non-incident participants. In the verification phase, high levels of MAT2β-AAb were significantly associated with the future incidence of KOA and with an earlier development of the disease. The incorporation of this AAb in a clinical model for the prognosis of incident radiographic KOA significantly improved the identification/classification of patients who will develop the disorder. The usefulness of the model to predict radiographic KOA was confirmed on a different OAI subcohort. Conclusions The measurement of AAbs against MAT2β in serum might be highly useful to improve the prediction of OA development, and also to estimate the time to incidence.
Collapse
Affiliation(s)
- María Camacho-Encina
- Grupo de Investigación de Reumatología, Unidad de Proteomica, INIBIC-Complejo Hospitalario Universitario A Coruña, SERGAS, Universidad de A Coruña, A Coruña, Spain
| | - Vanesa Balboa-Barreiro
- Grupo de Epidemiología Clínica y Bioestadística, INIBIC-Complejo Hospitalario Universitario A Coruña, SERGAS, Universidad de A Coruña, A Coruña, Spain
| | - Ignacio Rego-Perez
- Grupo de Investigacion de Reumatologia, Unidad de Genomica, INIBIC-Complexo Hospitalario Universitario de A Coruña, SERGAS, Universidad de A Coruña, A Coruña, Spain
| | - Florencia Picchi
- Grupo de Investigación de Reumatología, Unidad de Proteomica, INIBIC-Complejo Hospitalario Universitario A Coruña, SERGAS, Universidad de A Coruña, A Coruña, Spain
| | - Jennifer VanDuin
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute-Arizona State University, Tempe, Arizona, USA
| | - Ji Qiu
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute-Arizona State University, Tempe, Arizona, USA
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus. Proteomics Unit. CIBER-ONC, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), Salamanca, Spain
| | - Natividad Oreiro
- Grupo de Investigacion Reumatologia, Unidad de Investigacion Clinica, INIBIC-Complejo Hospitalario Universitario A Coruña, SERGAS, Universidad de A Coruña, A Coruña, Spain
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute-Arizona State University, Tempe, Arizona, USA
| | - Cristina Ruiz-Romero
- Grupo de Investigación de Reumatología, Unidad de Proteomica, INIBIC-Complejo Hospitalario Universitario A Coruña, SERGAS, Universidad de A Coruña, A Coruña, Spain
| | - Francisco J Blanco
- Grupo de Investigacion de Reumatologia, INIBIC-Complejo Hospitalario Universitario A Coruña, SERGAS, Departamento de Medicina, Universidad de A Coruña, A Coruña, Spain
| |
Collapse
|
30
|
Wang H, Huang Y, Bao P, Wu J, Zeng G, Hu X. β1,4-galactosyltransferase-I protects chondrocytes against TNF-induced apoptosis by blocking the TLR4 signaling pathway. Am J Transl Res 2019; 11:4358-4366. [PMID: 31396341 PMCID: PMC6684904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/13/2019] [Indexed: 06/10/2023]
Abstract
Osteoarthritis (OA) is the most common degenerative disease of the cartilage and is characterized by inflammation of the synovial membrane and subchondral osteosclerosis. β1,4-galactosyltransferase-I (β1,4-GalT-I) is a crucial regulator of inflammation based on its role in the stimulation and sustenance of inflammation in OA. In the present study, we aimed at elucidating the expression pattern and potential biological activity of β1,4-GalT-I in chondrocytes isolated from OA patients. Chondrocytes were isolated from the cartilage and cultured. Western blotting and quantitative real-time polymerase chain reaction (qRT-PCR) were used to analyze β1,4-GalT-I expression. Isolated chondrocytes were stimulated with tumor necrosis factor (TNF). Our results indicate significantly enhanced expression of β1,4-GalT-I in cultivated chondrocytes upon stimulation with TNF. β1,4-GalT-I inhibited the inflammation and cell death triggered by TNF. In addition, β1,4-GalT-Iinhibited the expression of Toll-like receptor 4 (TLR4) and phosphorylation of p65 and IKK. In conclusion, our findings suggest the protective effect of β1,4-GalT-I in chondrocytes against OA induced by TNF based on its ability to block the TLR4 signaling pathway. Our results also indicate significant contribution of β1,4-GalT-I towards the anti-inflammation in the cartilage of patients suffering from OA.
Collapse
Affiliation(s)
- Hairong Wang
- Department of Orthopedics, Jianhu Hospital Affiliated to Nantong UniversityYancheng, Jiangsu, China
| | - Yi Huang
- Department of Orthopedics, Jianhu Hospital Affiliated to Nantong UniversityYancheng, Jiangsu, China
| | - Peng Bao
- Department of Medical, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Jionglin Wu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Gang Zeng
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou, Guangdong, China
| | - Xumin Hu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou, Guangdong, China
| |
Collapse
|
31
|
Duchi S, Doyle S, Eekel T, D O'Connell C, Augustine C, Choong P, Onofrillo C, Di Bella C. Protocols for Culturing and Imaging a Human Ex Vivo Osteochondral Model for Cartilage Biomanufacturing Applications. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E640. [PMID: 30791632 PMCID: PMC6416585 DOI: 10.3390/ma12040640] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/01/2023]
Abstract
Cartilage defects and diseases remain major clinical issues in orthopaedics. Biomanufacturing is now a tangible option for the delivery of bioscaffolds capable of regenerating the deficient cartilage tissue. However, several limitations of in vitro and experimental animal models pose serious challenges to the translation of preclinical findings into clinical practice. Ex vivo models are of great value for translating in vitro tissue engineered approaches into clinically relevant conditions. Our aim is to obtain a viable human osteochondral (OC) model to test hydrogel-based materials for cartilage repair. Here we describe a detailed step-by-step framework for the generation of human OC plugs, their culture in a perfusion device and the processing procedures for histological and advanced microscopy imaging. Our ex vivo OC model fulfils the following requirements: the model is metabolically stable for a relevant culture period of 4 weeks in a perfusion bioreactor, the processing procedures allowed for the analysis of 3 different tissues or materials (cartilage, bone and hydrogel) without compromising their integrity. We determined a protocol and the settings for a non-linear microscopy technique on label free sections. Furthermore, we established a clearing protocol to perform light sheet-based observations on the cartilage layer without the need for tedious and destructive histological procedures. Finally, we showed that our OC system is a clinically relevant in terms of cartilage regeneration potential. In conclusion, this OC model represents a valuable preclinical ex vivo tool for studying cartilage therapies, such as hydrogel-based bioscaffolds, and we envision it will reduce the number of animals needed for in vivo testing.
Collapse
Affiliation(s)
- Serena Duchi
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
| | - Stephanie Doyle
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- School of Engineering, Discipline of Electrical and Biomedical Engineering, RMIT University, 124 La Trobe Street, 3000 Melbourne, Australia.
| | - Timon Eekel
- University of Utrecht, Domplein 29, 3512 JE Utrecht, The Netherlands.
| | - Cathal D O'Connell
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
| | - Cheryl Augustine
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
| | - Peter Choong
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- Department of Orthopaedics, St Vincent's Hospital, 41 Victoria Parade, 3065 Fitzroy, Australia.
| | - Carmine Onofrillo
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
| | - Claudia Di Bella
- BioFab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- Department of Surgery, St Vincent's Hospital, University of Melbourne, Clinical Sciences Building, 29 Regent Street, 3065 Fitzroy, Australia.
- Department of Orthopaedics, St Vincent's Hospital, 41 Victoria Parade, 3065 Fitzroy, Australia.
| |
Collapse
|