1
|
Han G, Cai L, Li B, Li Q, Yue luo, Wang Q, Kang P. Bibliometric analysis of synovial in osteoarthritis in the last 10 years. Heliyon 2024; 10:e33406. [PMID: 39035546 PMCID: PMC11259837 DOI: 10.1016/j.heliyon.2024.e33406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024] Open
Abstract
Background Our aim was to examine trends in the bibliometric analysis of synovial for osteoarthritis over the last 10 years. Methods Publications relevant to synovial in osteoarthritis from 2013 to 2022 were retrieved from the Science Citation Index Expanded (SCI-E), Social Sciences Citation Index (SSCI), and Web of Science Core Collection (WoSCC) databases. The countries/regions, institutions, authors, journals, references, and keywords related to this topic were extracted using Citespace and Vosviewer. Citespace and Vosviewer were also used to identify and analyze this field's research hotspots and trends. Results Over the past 10 years, 5738 articles addressing the role of synovium in osteoarthritis have been published. Between 2013 and 2022, 2021 had the highest amount of published articles (a total of 756 published articles, or 13.18 % of the total articles) covering synovial in osteoarthritis. China was the country that published the most articles, while Duke University was the institution that published the most articles. Osteoarthritis and Cartilage was the journal with the most publications related to the study of Synovium in osteoarthritis. The National Nature Science Foundation of China provided the most funding. According to the analysis of keyword burst detection, human cartilage, control experiment, and exosomes were the most searched at different points in time. Conclusion In the last ten years, both the number of citations and the article discussing synovial in osteoarthritis have increased. The top 10 most searched keywords were "osteoarthritis","synovial fluid", "inflammation", "cartilage", "expression","rheumatoid arthritis","articular cartilage", "knee osteoarthritis", "synovial", "knee". According to the timeline view of co-citation clustering, synovial components and their expressions have emerged as hotspots of research associated with synovial osteoarthritis.
Collapse
Affiliation(s)
- Guangtao Han
- West China Hospital of Sichuan University, China
| | - Lijun Cai
- West China Hospital of Sichuan University, China
| | - Bohua Li
- West China Hospital of Sichuan University, China
| | - Qianhao Li
- West China Hospital of Sichuan University, China
| | - Yue luo
- West China Hospital of Sichuan University, China
| | - Qiuru Wang
- West China Hospital of Sichuan University, China
| | - Pengde Kang
- West China Hospital of Sichuan University, China
| |
Collapse
|
2
|
St Amant J, Michaud J, Hinds D, Coyle M, Pozzi A, Clark AL. Depleting transforming growth factor beta receptor 2 signalling in the cartilage of itga1-null mice attenuates spontaneous knee osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2023; 5:100399. [PMID: 37649532 PMCID: PMC10462827 DOI: 10.1016/j.ocarto.2023.100399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
Objectives Integrin α1β1 protects against osteoarthritis (OA) when it is upregulated in the superficial zone of cartilage in the early stages of disease. However, the mechanism behind this protection is unknown. Integrin α1β1 moderates transforming growth factor β receptor II (TGFBR2) signalling, a critical regulator of chondrocyte anabolic activity. To this end, mice lacking integrin α1β1 have increased baseline activation of TGFBR2 signalling and overall fibrosis. The purpose of this study was to evaluate the interplay between integrin α1β1 and TGFBR2 in the development of spontaneous OA. We hypothesized that dampening TGFBR2 signalling in the cartilage of itga1-null mice would attenuate OA. Methods Behavioural and histological manifestations of spontaneous knee OA were measured at 4, 8, 12 and 16 months in mice with and without a ubiquitous itga1 deletion and with and without a tamoxifen-induced cartilage specific TGFBR2 depletion. Results Knee cartilage degeneration, collateral ligament ossification and pain responses increased with age. Itga1-null mice with intact TGFBR2 signalling developed earlier and more severe OA compared to controls. In agreement with our hypothesis, depleting TGFBR2 signalling in the cartilage of itga1-null mice attenuated OA progression. Conclusion Intact TGFBR2 signalling drives early and worse knee OA in itga1-null mice. This result supports the hypothesis that the increased expression of integrin α1β1 by superficial zone chondrocytes early in OA development dampens TGFBR2 signalling and thus protects against degeneration.
Collapse
Affiliation(s)
- Jennifer St Amant
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Jana Michaud
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Daniel Hinds
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Madison Coyle
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Andrea L. Clark
- Human Health and Nutritional Sciences, College of Biological Science, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
3
|
Yilmaz M, Dokuyucu R. Effects of Thymoquinone on Urotensin-II and TGF-β1 Levels in Model of Osteonecrosis in Rats. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1781. [PMID: 37893499 PMCID: PMC10608466 DOI: 10.3390/medicina59101781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023]
Abstract
Objectives: We aimed to investigate the therapeutic effects of thymoquinone (TMQ) treatment in osteonecrotic rats by evaluating protein levels, osteonecrosis (ON) levels, fatty acid degeneration, oxidative status, and plasma levels of Urotensin-II (U-II) and transforming growth factor-beta (TGF-β1). Materials and Methods: 40 weight-matched adult male Wistar rats were grouped as control (n = 10), methylprednisolone acetate (MPA) (n = 10), thymoquinone (TMQ) (n = 10), and MPA + TMQ (n = 10). To induce ON, 15-week-old animals were subcutaneously injected with MPA at a dose of 15 mg/kg twice weekly for 2 weeks. TMQ was injected into 15-week-old rats via gastric gavage at a dose of 80 mg/kg per day for 4 weeks. The rats in the MPA + TMQ group were administered TMQ 2 weeks before the MPA injection. At the end of the treatments, cardiac blood samples and femur samples were collected for biochemical and histological evaluations. Results: In the control and TMQ groups, no ON pattern was observed. However, in tissues exposed to MPA, TMQ treatment resulted in significantly decreased ON levels compared to the MPA group. The number of cells that were positive for 8-OHdG and 4-HNE was significantly lower in the MPA + TMQ group than in the MPA group (p < 0.05). In terms of TGF-β1 and U-II levels, we observed that both TGF-β1 (367.40 ± 23.01 pg/mL vs. 248.9 ± 20.12 pg/mL) and U-II protein levels (259.5 ± 6.0 ng/mL vs. 168.20 ± 7.90 ng/mL) increased significantly in the MPA group compared to the control group (p < 0.001). Furthermore, TGF-β1 (293.50 ± 14.18 pg/mL) and U-II (174.80 ± 4.2 ng/mL) protein levels were significantly decreased in the MPA + TMQ group compared to the MPA group (p < 0.05 and p < 0.01, respectively). There was a statistically positive correlation (p < 0.05) between the TGF-β1 and U-II protein levels in all groups (p = 0.002, rcontrol = 0.890; p = 0.02, rTMQ = 0.861; p = 0.024, rMPA+TMQ = 0.868) except for the MPA group (p < 0.03, rMedrol = -0.870). Conclusions: As far as we know, this is the first study to demonstrate the curative functions of TMQ on ON by causing a correlated decrease in the expression of U-II and TGF-β1 in the femoral heads of rats.
Collapse
Affiliation(s)
- Mehmet Yilmaz
- Department of Orthopedic Surgery, 25 Aralik State Hospital, Gaziantep 27090, Turkey;
| | - Recep Dokuyucu
- Department of Physiology, Private Fizyoclinic Wellness Center, Gaziantep 27560, Turkey
| |
Collapse
|
4
|
Liu F, Su F, Zhang T, Liu R, Liu N, Dong T. Relationship between knee osteophytes and calcitonin gene-related peptide concentrations of serum and synovial fluid in knee of osteoarthritis. Medicine (Baltimore) 2023; 102:e34691. [PMID: 37657066 PMCID: PMC10476791 DOI: 10.1097/md.0000000000034691] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/20/2023] [Indexed: 09/03/2023] Open
Abstract
To explore the relationship between knee osteophytes of osteoarthritic knee and calcitonin gene-related peptide (CGRP) concentrations of serum and synovial fluid (SF). 65 patients with knee medial compartment osteoarthritis (OA) were recruited and examined with weight-bearing radiographs of the entire lower limb. The concentrations of CGRP in serum/SF were also detected in surgery. The relationship between the concentrations of CGRP in serum/SF and osteophyte scores were detected with Spearman rank correlation coefficient. CGRP concentrations in serum and SF were significantly correlated with osteophyte score of overall knee respectively (R = 0.462, P < .001; R = 0.435, P < .001). In addition, a correlation tended to be observed about the relationship between CGRP concentrations in serum and SF and osteophyte scores of medial compartment (R = 0.426, P < .001; R = 0.363, P = .003), and osteophyte scores of lateral compartment (R = 0.429, P < .001; R = 0.444, P < .001). In this study, the relationship between CGRP in serum/SF and knee osteophyte scores in different subregions were explored, which showed significant positive correlations, that possibly reflecting the contribution of CGRP influencing osteophyte formation. Positive correlations between osteophyte scores and CGRP suggest that CGRP promote the growth of osteophyte formation. It has the potential to be selected as a biomarker for the assessment of severity in knee OA patients and predict the progression of knee OA. It also provides a potential therapeutic target to delay the progression and relieve the symptom of OA.
Collapse
Affiliation(s)
- Fan Liu
- Department of Orthopaedic Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Fan Su
- Department of Orthopaedic Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Tao Zhang
- Department of Orthopaedic Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Rui Liu
- Department of Orthopaedic Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Na Liu
- Department of Orthopaedic Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Tianhua Dong
- Department of Orthopaedic Surgery, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| |
Collapse
|
5
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
6
|
MRI underestimates presence and size of knee osteophytes using CT as a reference standard. Osteoarthritis Cartilage 2023; 31:656-668. [PMID: 36796577 DOI: 10.1016/j.joca.2023.01.575] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/12/2023] [Accepted: 01/31/2023] [Indexed: 02/18/2023]
Abstract
OBJECTIVE To explore the diagnostic performance of routine magnetic resonance imaging (MRI) for the cross-sectional assessment of osteophytes (OPs) in all three knee compartments using computed tomography (CT) as a reference standard. METHODS The Strontium Ranelate Efficacy in Knee Osteoarthritis (SEKOIA) trial explored the effect of 3 years of treatment with strontium ranelate in patients with primary knee OA. OPs were scored for the baseline visit only using a modified MRI Osteoarthritis Knee Score (MOAKS) scoring system in the patellofemoral (PFJ), the medial tibiofemoral (TFJ) and the lateral TFJ. Size was assessed from 0 to 3 in 18 locations. Descriptive statistics were used to describe differences in ordinal grading between CT and MRI. In addition, weighted-kappa statistics were employed to assess agreement between scoring using the two methods. Sensitivity, specificity, positive predictive value and negative predictive value as well as area under the curve (AUC) measures of diagnostic performance were employed using CT as the reference standard. RESULTS Included were 74 patients with available MRI and CT data. Mean age was 62.9 ± 7.5 years. Altogether 1,332 locations were evaluated. For the PFJ, MRI detected 141 (72%) of 197 CT-defined OPs with a w-kappa of 0.58 (95% CI [0.52-0.65]). In the medial TFJ, MRI detected 178 (81%) of 219 CT-OPs with a w-kappa of 0.58 (95% CI [0.51-0.64]). For the lateral compartment these numbers were 84 (70%) of 120 CT-OPs with a w-kappa of 0.58 (95% CI [0.50-0.66]). CONCLUSION MRI underestimates presence of osteophytes in all three knee compartments. CT may be helpful particularly regarding assessment of small osteophytes particularly in early disease.
Collapse
|
7
|
Markhardt BK, Huang BK, Spiker AM, Chang EY. Interpretation of Cartilage Damage at Routine Clinical MRI: How to Match Arthroscopic Findings. Radiographics 2022; 42:1457-1473. [PMID: 35984752 PMCID: PMC9453290 DOI: 10.1148/rg.220051] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/11/2022]
Abstract
This review is intended to aid in the interpretation of damage to the articular cartilage at routine clinical MRI to improve clinical management. Relevant facets of the histologic and biochemical characteristics and clinical management of cartilage are discussed, as is MRI physics. Characterization of damage to the articular cartilage with MRI demands a detailed understanding of the normal and damaged appearance of the osteochondral unit in the context of different sequence parameters. Understanding the location of the subchondral bone plate is key to determining the depth of the cartilage lesion. Defining the bone plate at MRI is challenging because of the anisotropic fibrous organization of articular cartilage, which is susceptible to the "magic angle" phenomenon and chemical shift artifacts at the interface with the fat-containing medullary cavity. These artifacts may cause overestimation of the thickness of the subchondral bone plate and, therefore, overestimation of the depth of a cartilage lesion. In areas of normal cartilage morphology, isolated hyperintense and hypointense lesions often represent degeneration of cartilage at arthroscopy. Changes in the subchondral bone marrow at MRI also increase the likelihood that cartilage damage will be visualized at arthroscopy, even when a morphologic lesion cannot be resolved, and larger subchondral lesions are associated with higher grades at arthroscopy. The clinical significance of other secondary features of cartilage damage are also reviewed, including osteophytes, intra-articular bodies, and synovitis. Online supplemental material is available for this article. Work of the U.S. Government published under an exclusive license with the RSNA.
Collapse
Affiliation(s)
- B. Keegan Markhardt
- From the Department of Radiology, Division of Musculoskeletal Imaging
and Intervention (B.K.M.), and Department of Orthopedic Surgery (A.M.S.),
University of Wisconsin-Madison, Clinical Science Center, 600 Highland Ave,
E3/311, Madison, WI 53792; Department of Radiology, Division of Musculoskeletal
Imaging, University of California, San Diego, La Jolla, Calif (B.K.H., E.Y.C.);
and Radiology Services, Veterans Affairs San Diego Healthcare System
(E.Y.C.)
| | - Brady K. Huang
- From the Department of Radiology, Division of Musculoskeletal Imaging
and Intervention (B.K.M.), and Department of Orthopedic Surgery (A.M.S.),
University of Wisconsin-Madison, Clinical Science Center, 600 Highland Ave,
E3/311, Madison, WI 53792; Department of Radiology, Division of Musculoskeletal
Imaging, University of California, San Diego, La Jolla, Calif (B.K.H., E.Y.C.);
and Radiology Services, Veterans Affairs San Diego Healthcare System
(E.Y.C.)
| | - Andrea M. Spiker
- From the Department of Radiology, Division of Musculoskeletal Imaging
and Intervention (B.K.M.), and Department of Orthopedic Surgery (A.M.S.),
University of Wisconsin-Madison, Clinical Science Center, 600 Highland Ave,
E3/311, Madison, WI 53792; Department of Radiology, Division of Musculoskeletal
Imaging, University of California, San Diego, La Jolla, Calif (B.K.H., E.Y.C.);
and Radiology Services, Veterans Affairs San Diego Healthcare System
(E.Y.C.)
| | - Eric Y. Chang
- From the Department of Radiology, Division of Musculoskeletal Imaging
and Intervention (B.K.M.), and Department of Orthopedic Surgery (A.M.S.),
University of Wisconsin-Madison, Clinical Science Center, 600 Highland Ave,
E3/311, Madison, WI 53792; Department of Radiology, Division of Musculoskeletal
Imaging, University of California, San Diego, La Jolla, Calif (B.K.H., E.Y.C.);
and Radiology Services, Veterans Affairs San Diego Healthcare System
(E.Y.C.)
| |
Collapse
|
8
|
Terzi MY, Okuyan HM, Karaboğa İ, Gökdemir CE, Tap D, Kalacı A. Urotensin-II Prevents Cartilage Degeneration in a Monosodium Iodoacetate-Induced Rat Model of Osteoarthritis. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10448-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
9
|
McKinney JM, Pucha KA, Doan TN, Wang L, Weinstock LD, Tignor BT, Fowle KL, Levit RD, Wood LB, Willett NJ. Sodium alginate microencapsulation of human mesenchymal stromal cells modulates paracrine signaling response and enhances efficacy for treatment of established osteoarthritis. Acta Biomater 2022; 141:315-332. [PMID: 34979327 DOI: 10.1016/j.actbio.2021.12.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 01/15/2023]
Abstract
Mesenchymal stromal cells (MSCs) have shown promise as osteoarthritis (OA) treatments; however, effective translation has been limited by high variability and heterogeneity of MSCs, suboptimal delivery strategies, and poor understanding of critical quality and potency attributes. Furthermore, most pre-clinical studies of MSC therapeutics for OA have focused on delaying OA development and not on treating established OA, which brings added clinical relevance. Thus, the objective of the current study was to assess the effects of sodium alginate microencapsulation on human MSC (hMSC) secretion of immunomodulatory cytokines in an OA microenvironment and therapeutic efficacy in treating established OA. A Medial Meniscal Transection (MMT) pre-clinical model of OA was implemented. Three weeks post-surgery, after OA was established, intra-articular injections of encapsulated hMSCs or nonencapsulated hMSCs were administered. Six weeks post-surgery, microstructural changes in the knee joint were quantified using microCT. Encapsulated hMSCs reduced articular cartilage degeneration and subchondral bone remodeling. A multiplexed immunoassay panel was used to profile the in vitro secretome of hMSCs in response to IL-1β. Nonencapsulated hMSCs showed an indiscriminate increase in all cytokines in response to IL-1β while encapsulated hMSCs showed a targeted secretory response with increased expression of pro-inflammatory (IL-1β, IL-6, IL-7, IL-8), anti-inflammatory (IL-1RA), and chemotactic (G-CSF, MDC, IP10) cytokines. These data show that sodium alginate microencapsulation can modulate hMSC paracrine signaling and enhance the therapeutic efficacy of the hMSCs in treating established OA. This cytokine profile provides a foundation for the identification of key factors affecting the overall potency of hMSC therapeutics for OA. STATEMENT OF SIGNIFICANCE: While there has been considerable interest in material based MSC encapsulation for treatment of OA, there are critical gaps in our translational understanding of these biomaterial-based technologies for OA. More specifically, previous studies have several important limitations: (1) they have been largely focused on preventing OA development, which limits their translational utility and (2) little prior work has been done to delineate potential routes/mechanisms by which material encapsulation alters MSC therapeutic action. In our manuscript, we aimed to fill these gaps in knowledge by testing the hypotheses that: (1) hMSC encapsulation can attenuate established disease progression, which is a more clinically relevant scenario and (2) hMSC encapsulation significantly changes the secreted paracrine factors from hMSCs.
Collapse
Affiliation(s)
- Jay M McKinney
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA; Department of Orthopaedics, Emory University, 49 Jesse Hill Jr Dr SE, Atlanta, GA 30303, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Krishna A Pucha
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA
| | - Thanh N Doan
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA; Department of Orthopaedics, Emory University, 49 Jesse Hill Jr Dr SE, Atlanta, GA 30303, USA
| | - Lanfang Wang
- Department of Medicine, Division of Cardiology, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Laura D Weinstock
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Benjamin T Tignor
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Kelsey L Fowle
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA
| | - Rebecca D Levit
- Department of Medicine, Division of Cardiology, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, North Ave NW, Atlanta, GA 30332, USA.
| | - Nick J Willett
- Research Division, VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, USA; Department of Orthopaedics, Emory University, 49 Jesse Hill Jr Dr SE, Atlanta, GA 30303, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA 30332, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA 30332, USA; Phil and Penny Knight Campus for Accelerating Scientific Impact, 6231 University of Oregon, Eugene, Oregon, USA.
| |
Collapse
|
10
|
van der Kraan PM. Inhibition of transforming growth factor-β in osteoarthritis. Discrepancy with reduced TGFβ signaling in normal joints. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100238. [PMID: 36474474 PMCID: PMC9718219 DOI: 10.1016/j.ocarto.2022.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 10/19/2022] Open
Abstract
Objective Transforming growth factor-β (TGFβ) is a pleiotropic cytokine that is central in the regulation of joint health and disease. Inhibition of TGFβ activity/signaling in experimental osteoarthritis (OA) has been performed to modulate OA severity and progression. In this narrative review we discuss the potential reasons for the variable results of TGFβ inhibition in these models. Design A literature study was performed using the search terms; experimental osteoarthritis and TGFβ. Papers were selected that describe the effect TGFβ activity/signaling inhibition on experimental OA. Based on the selected papers a narrative review has been written about the potential therapeutic role of TGFβ inhibition in OA and potential causes for its variable effects are discussed. Results Inhibition of TGFβ activity in experimental models of OA does not result in either straightforward protection or deleterious effects. More than half of the studies (13/19), but not all, report that inhibition of TGFβ in experimental OA reduces OA severity. This is in contrast with the protective role of TGFβ in healthy joints. Conclusions The effect of TGFβ inhibition on joint damage in experimental OA is variable. Most likely this is a consequence of the changing function of TGFβ in normal and OA joints. As a result, the overall outcome of TGFβ modulation in OA will be unpredictable. To develop OA therapies based on modulation of TGFβ activity specific protective and damaging signaling routes should be identified and tools developed to block the damaging ones.
Collapse
|
11
|
Mast Cells Differentiated in Synovial Fluid and Resident in Osteophytes Exalt the Inflammatory Pathology of Osteoarthritis. Int J Mol Sci 2022; 23:ijms23010541. [PMID: 35008966 PMCID: PMC8745477 DOI: 10.3390/ijms23010541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/26/2021] [Accepted: 12/28/2021] [Indexed: 02/01/2023] Open
Abstract
Introduction: Osteophytes are a prominent feature of osteoarthritis (OA) joints and one of the clinical hallmarks of the disease progression. Research on osteophytes is fragmentary and modes of its contribution to OA pathology are obscure. Aim: To elucidate the role of osteophytes in OA pathology from a perspective of molecular and cellular events. Methods: RNA-seq of fully grown osteophytes, collected from tibial plateau of six OA patients revealed patterns corresponding to active extracellular matrix re-modulation and prominent participation of mast cells. Presence of mast cells was further confirmed by immunohistochemistry, performed on the sections of the osteophytes using anti-tryptase alpha/beta-1 and anti-FC epsilon RI antibodies and the related key up-regulated genes were validated by qRT-PCR. To test the role of OA synovial fluid (SF) in mast cell maturation as proposed by the authors, hematopoietic stem cells (HSCs) and ThP1 cells were cultured in a media supplemented with 10% SF samples, obtained from various grades of OA patients and were monitored using specific cell surface markers by flow cytometry. Proteomics analysis of SF samples was performed to detect additional markers specific to mast cells and inflammation that drive the cell differentiation and maturation. Results: Transcriptomics of osteophytes revealed a significant upregulation of mast cells specific genes such as chymase 1 (CMA1; 5-fold) carboxypeptidase A3 (CPA3; 4-fold), MS4A2/FCERI (FCERI; 4.2-fold) and interleukin 1 receptor-like 1 (IL1RL1; 2.5-fold) indicating their prominent involvement. (In IHC, anti-tryptase alpha/beta-1 and anti- FC epsilon RI-stained active mast cells were seen populated in cartilage, subchondral bone, and trabecular bone.) Based on these outcomes and previous learnings, the authors claim a possibility of mast cells invasion into osteophytes is mediated by SF and present in vitro cell differentiation assay results, wherein ThP1 and HSCs showed differentiation into HLA-DR+/CD206+ and FCERI+ phenotype, respectively, after exposing them to medium containing 10% SF for 9 days. Proteomics analysis of these SF samples showed an accumulation of mast cell-specific inflammatory proteins. Conclusions: RNA-seq analysis followed by IHC study on osteophyte samples showed a population of mast cells resident in them and may further accentuate inflammatory pathology of OA. Besides subchondral bone, the authors propose an alternative passage of mast cells invasion in osteophytes, wherein OA SF was found to be necessary and sufficient for maturation of mast cell precursor into effector cells.
Collapse
|
12
|
Weng PW, Yadav VK, Pikatan NW, Fong IH, Lin IH, Yeh CT, Lee WH. Novel NFκB Inhibitor SC75741 Mitigates Chondrocyte Degradation and Prevents Activated Fibroblast Transformation by Modulating miR-21/GDF-5/SOX5 Signaling. Int J Mol Sci 2021; 22:11082. [PMID: 34681754 PMCID: PMC8538686 DOI: 10.3390/ijms222011082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/04/2021] [Accepted: 10/09/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is a common articular disease manifested by the destruction of cartilage and compromised chondrogenesis in the aging population, with chronic inflammation of synovium, which drives OA progression. Importantly, the activated synovial fibroblast (AF) within the synovium facilitates OA through modulating key molecules, including regulatory microRNAs (miR's). To understand OA associated pathways, in vitro co-culture system, and in vivo papain-induced OA model were applied for this study. The expression of key inflammatory markers both in tissue and blood plasma were examined by qRT-PCR, western blot, immunohistochemistry, enzyme-linked immunosorbent assay (ELISA) and immunofluorescence assays. Herein, our result demonstrated, AF-activated human chondrocytes (AC) exhibit elevated NFκB, TNF-α, IL-6, and miR-21 expression as compared to healthy chondrocytes (HC). Importantly, AC induced the apoptosis of HC and inhibited the expression of chondrogenesis inducers, SOX5, TGF-β1, and GDF-5. NFκB is a key inflammatory transcription factor elevated in OA. Therefore, SC75741 (an NFκB inhibitor) therapeutic effect was explored. SC75741 inhibits inflammatory profile, protects AC-educated HC from apoptosis, and inhibits miR-21 expression, which results in the induced expression of GDF-5, SOX5, TGF-β1, BMPR2, and COL4A1. Moreover, ectopic miR-21 expression in fibroblast-like activated chondrocytes promoted osteoblast-mediated differentiation of osteoclasts in RW264.7 cells. Interestingly, in vivo study demonstrated SC75741 protective role, in controlling the destruction of the articular joint, through NFκB, TNF-α, IL-6, and miR-21 inhibition, and inducing GDF-5, SOX5, TGF-β1, BMPR2, and COL4A1 expression. Our study demonstrated the role of NFκB/miR-21 axis in OA progression, and SC75741's therapeutic potential as a small-molecule inhibitor of miR-21/NFκB-driven OA progression.
Collapse
Affiliation(s)
- Pei-Wei Weng
- Department of Orthopaedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan;
- Department of Orthopaedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City 110, Taiwan
| | - Vijesh Kumar Yadav
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; (V.K.Y.); (N.W.P.); (I.-H.F.); (C.-T.Y.)
| | - Narpati Wesa Pikatan
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; (V.K.Y.); (N.W.P.); (I.-H.F.); (C.-T.Y.)
| | - Iat-Hang Fong
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; (V.K.Y.); (N.W.P.); (I.-H.F.); (C.-T.Y.)
| | - I-Hsin Lin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei City 110, Taiwan
- Division of Periodontics, Department of Dentistry, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan
| | - Chi-Tai Yeh
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; (V.K.Y.); (N.W.P.); (I.-H.F.); (C.-T.Y.)
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu City 30015, Taiwan
| | - Wei-Hwa Lee
- Department of Medical Research & Education, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan; (V.K.Y.); (N.W.P.); (I.-H.F.); (C.-T.Y.)
- Department of Pathology, Taipei Medical University-Shuang Ho Hospital, New Taipei City 23561, Taiwan
| |
Collapse
|
13
|
Maglaviceanu A, Wu B, Kapoor M. Fibroblast-like synoviocytes: Role in synovial fibrosis associated with osteoarthritis. Wound Repair Regen 2021; 29:642-649. [PMID: 34021514 DOI: 10.1111/wrr.12939] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/02/2021] [Accepted: 05/08/2021] [Indexed: 12/12/2022]
Abstract
The synovial membrane undergoes a variety of structural changes throughout the pathogenesis of osteoarthritis (OA), including the development of fibrosis. Fibroblast-like synoviocytes (FLS) are a heterogenous cell population of the synovium that are suggested to drive the fibrotic response, but the exact mechanisms associated with their activation in OA remain unclear. Once activated, FLS are suggested to acquire a myofibroblast-like phenotype that drives fibrogenesis through excessive extracellular matrix (ECM) component deposition and an enhanced contractile function. In this review, we define FLS in the synovium, discuss how select extracellular or endogenous factors potentially induce their activation in OA, and describe how the activity of myofibroblast-like cells affects the structure of the synovial membrane.
Collapse
Affiliation(s)
- Anca Maglaviceanu
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Brian Wu
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Amin LE, Gamily ME. Biological impact of curcumin on the healing of tempromandibular joint in experimentally induced arthritis. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY, MEDICINE, AND PATHOLOGY 2021. [DOI: 10.1016/j.ajoms.2020.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
15
|
Cherifi C, Monteagudo S, Lories RJ. Promising targets for therapy of osteoarthritis: a review on the Wnt and TGF-β signalling pathways. Ther Adv Musculoskelet Dis 2021; 13:1759720X211006959. [PMID: 33948125 PMCID: PMC8053758 DOI: 10.1177/1759720x211006959] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disorder worldwide, with a high personal burden for the patients and an important socio-economic impact. Current therapies are largely limited to pain management and rehabilitation and exercise strategies. For advanced cases, joint replacement surgery may be the only option. Hence, there is an enormous need for the development of effective and safe disease-modifying anti-OA drugs. A strong focus in OA research has been on the identification and role of molecular signalling pathways that contribute to the balance between anabolism and catabolism in the articular cartilage. In this context, most insights have been gained in understanding the roles of the transforming growth factor-beta (TGF-β) and the Wingless-type (Wnt) signalling cascades. The emerging picture demonstrates a high degree of complexity with context-dependent events. TGF-β appears to protect cartilage under healthy conditions, but shifts in its receptor use and subsequent downstream signalling may be deleterious in aged individuals or in damaged cartilage. Likewise, low levels of Wnt activity appear important to sustain chondrocyte viability but excessive activation is associated with progressive joint damage. Emerging clinical data suggest some potential for the use of sprifermin, a recombinant forms of fibroblast growth factor 18, a distant TGF-β superfamily member, and for lorecivivint, a Wnt pathway modulator.
Collapse
Affiliation(s)
- Chahrazad Cherifi
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, Leuven, Belgium
| | - Silvia Monteagudo
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, Leuven, Belgium
| | - Rik J Lories
- Department of Development and Regeneration, KU Leuven, Skeletal Biology and Engineering Research Centre, Box 813 O&N, Herestraat 49, Leuven 3000, Belgium; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Reed KSM, Ulici V, Kim C, Chubinskaya S, Loeser RF, Phanstiel DH. Transcriptional response of human articular chondrocytes treated with fibronectin fragments: an in vitro model of the osteoarthritis phenotype. Osteoarthritis Cartilage 2021; 29:235-247. [PMID: 33248223 PMCID: PMC7870543 DOI: 10.1016/j.joca.2020.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/19/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Fibronectin is a matrix protein that is fragmented during cartilage degradation in osteoarthritis (OA). Treatment of chondrocytes with fibronectin fragments (FN-f) has been used to model OA in vitro, but the system has not been fully characterized. This study sought to define the transcriptional response of chondrocytes to FN-f, and directly compare it to responses traditionally observed in OA. DESIGN Normal human femoral chondrocytes isolated from tissue donors were treated with either FN-f or PBS (control) for 3, 6, or 18 h. RNA-seq libraries were compared between time-matched FN-f and control samples in order to identify changes in gene expression over time. Differentially expressed genes were compared to a published OA gene set and used for pathway, transcription factor motif, and kinome analysis. RESULTS FN-f treatment resulted in 3,914 differentially expressed genes over the time course. Genes that are up- or downregulated in OA were significantly up- (P < 0.00001) or downregulated (P < 0.0004) in response to FN-f. Early response genes were involved in proinflammatory pathways, whereas many late response genes were involved in ferroptosis. The promoters of upregulated genes were enriched for NF-κB, AP-1, and IRF motifs. Highly upregulated kinases included CAMK1G, IRAK2, and the uncharacterized kinase DYRK3, while growth factor receptors TGFBR2 and FGFR2 were downregulated. CONCLUSIONS FN-f treatment of normal human articular chondrocytes recapitulated many key aspects of the OA chondrocyte phenotype. This in vitro model is promising for future OA studies, especially considering its compatibility with genomics and genome-editing techniques.
Collapse
Affiliation(s)
- K S M Reed
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - V Ulici
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - C Kim
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - S Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| | - R F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| | - D H Phanstiel
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA; Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
17
|
Rim YA, Ju JH. The Role of Fibrosis in Osteoarthritis Progression. Life (Basel) 2020; 11:life11010003. [PMID: 33374529 PMCID: PMC7822172 DOI: 10.3390/life11010003] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease where the main characteristics include cartilage degeneration and synovial membrane inflammation. These changes in the knee joint eventually dampen the function of the joint and restrict joint movement, which eventually leads to a stage where total joint replacement is the only treatment option. While much is still unknown about the pathogenesis and progression mechanism of OA, joint fibrosis can be a critical issue for better understanding this disease. Synovial fibrosis and the generation of fibrocartilage are the two main fibrosis-related characteristics that can be found in OA. However, these two processes remain mostly misunderstood. In this review, we focus on the fibrosis process in OA, especially in the cartilage and the synovium tissue, which are the main tissues involved in OA.
Collapse
Affiliation(s)
- Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-6895
| |
Collapse
|
18
|
Tryfonidou MA, de Vries G, Hennink WE, Creemers LB. "Old Drugs, New Tricks" - Local controlled drug release systems for treatment of degenerative joint disease. Adv Drug Deliv Rev 2020; 160:170-185. [PMID: 33122086 DOI: 10.1016/j.addr.2020.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA) and chronic low back pain (CLBP) caused by intervertebral disc (IVD) degeneration are joint diseases that have become major causes for loss of quality of life worldwide. Despite the unmet need, effective treatments other than invasive, and often ineffective, surgery are lacking. Systemic administration of drugs entails suboptimal local drug exposure in the articular joint and IVD. This review provides an overview of the potency of biomaterial-based drug delivery systems as novel treatment modality, with a focus on the biological effects of drug release systems that have reached translation at the level of in vivo models and relevant ex vivo models. These studies have shown encouraging results of biomaterial-based local delivery of several types of drugs, mostly inhibitors of inflammatory cytokines or other degenerative factors. Prevention of inflammation and degeneration and pain relief was achieved, although mainly in small animal models, with interventions applied at an early disease stage. Less convincing data were obtained with the delivery of regenerative factors. Multidisciplinary efforts towards tackling the discord between in vitro and in vivo release, combined with adaptations in the regulatory landscape may be needed to enhance safe and expeditious introduction of more and more effective controlled release-based treatments with the OA and CLBP patients.
Collapse
|
19
|
Mobasheri A, Choi H, Martín-Vasallo P. Over-Production of Therapeutic Growth Factors for Articular Cartilage Regeneration by Protein Production Platforms and Protein Packaging Cell Lines. BIOLOGY 2020; 9:biology9100330. [PMID: 33050357 PMCID: PMC7599991 DOI: 10.3390/biology9100330] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022]
Abstract
Simple Summary Osteoarthritis (OA) is the most common form of arthritis across the world. Most of the existing drugs for OA treat the symptoms of pain and inflammation. There are no drugs that can dure the disease. There are a number of new treatments for OA including cell therapy and gene therapy. This articles outlines the concept behind TissueGene-C, a new biological drug for OA. This new treatment includes cartilage cells mixed with a genetically modified cell line called GP2-293, which is effectively a “drug factory”, over-producing the growth factors that are important for cartilage regeneration and changing the environment inside joints. The mixture is injected into the affected knee joint. These cells are designed to be short-lived and cannot reproduce. Therefore, after they have done their job, they die and are cleared by immune cells. This is a new and modern approach to treating OA and TissueGene-C is the prototype cell therapy for OA. In the future, it is entirely possible to combine different clones of genetically engineered cells like GP2-293 that have been designed to over-produce a growth factor or biological drug with cells from the cartilage endplate of the intervertebral disc to treat degeneration in the spine. Abstract This review article focuses on the current state-of-the-art cellular and molecular biotechnology for the over-production of clinically relevant therapeutic and anabolic growth factors. We discuss how the currently available tools and emerging technologies can be used for the regenerative treatment of osteoarthritis (OA). Transfected protein packaging cell lines such as GP-293 cells may be used as “cellular factories” for large-scale production of therapeutic proteins and pro-anabolic growth factors, particularly in the context of cartilage regeneration. However, when irradiated with gamma or x-rays, these cells lose their capacity for replication, which makes them safe for use as a live cell component of intra-articular injections. This innovation is already here, in the form of TissueGene-C, a new biological drug that consists of normal allogeneic primary chondrocytes combined with transduced GP2-293 cells that overexpress the growth factor transforming growth factor β1 (TGF-β1). TissueGene-C has revolutionized the concept of cell therapy, allowing drug companies to develop live cells as biological drug delivery systems for direct intra-articular injection of growth factors whose half-lives are in the order of minutes. Therefore, in this paper, we discuss the potential for new innovations in regenerative medicine for degenerative diseases of synovial joints using mammalian protein production platforms, specifically protein packaging cell lines, for over-producing growth factors for cartilage tissue regeneration and give recent examples. Mammalian protein production platforms that incorporate protein packaging eukaryotic cell lines are superior to prokaryotic bacterial expression systems and are likely to have a significant impact on the development of new humanized biological growth factor therapies for treating focal cartilage defects and more generally for the treatment of degenerative joint diseases such as OA, especially when injected directly into the joint.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, FI-90014 Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Versus Arthritis Centre for Sport, Exercise and Osteoarthritis Research, Queen’s Medical Centre, Nottingham NG7 2UH, UK
- Correspondence: or
| | - Heonsik Choi
- Kolon TissueGene, Inc., Rockville, MD 20850, USA;
- Healthcare Research Institute, Kolon Advanced Research Center, Kolon Industries, Inc., Magok-dong, Gangseo-gu, Seoul 07793, Korea
| | - Pablo Martín-Vasallo
- UD of Biochemistry and Molecular Biology, Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, 38071 Tenerife, Spain;
| |
Collapse
|
20
|
Roux CH, Coste J, Roger C, Fontas E, Rat AC, Guillemin F. Impact of smoking on femorotibial and hip osteoarthritis progression: 3-year follow-up data from the KHOALA cohort. Joint Bone Spine 2020; 88:105077. [PMID: 32950705 DOI: 10.1016/j.jbspin.2020.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/08/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVES To evaluate the clinical and structural impact of smoking on knee and hip osteoarthritis at baseline and after 3years. METHODS Observational data on the progressive effects of smoking at baseline and after 3years were collected from The Knee and Hip Osteoarthritis Long-Term Assessment cohort comprising a French population of patients aged 40-75years with symptomatic lower limb osteoarthritis. Clinical (the Western Ontario and McMaster Universities Arthritis Index and Harris scores) and structural (radiography for osteophyte detection and joint-space narrowing assessment) were conducted. The tobacco usage categories were 'never smoker', 'former smoker', and 'current smoker'. RESULTS Of the 873 subjects included, 215 (25%) were former smokers and 119 (14%) were current smokers. Multivariate analyses revealed that former and current smokers had fewer knee osteophytes in the medial compartment at baseline (odds ratio [OR]=0.64 [0.41-0.99] and 0.63 [0.36-1.11], respectively), lower osteophyte development in the lateral condyle after 3years (OR=011 [0.03-0.45] and 0.15 [0.03-0.97]), and lower osteophyte development in the lateral tibial plateau after 3years (OR=0.22 [0.06-0.75] and 0.68 [0.14-3.35]). Higher tobacco consumption and longer duration of consumption were significantly associated with fewer knee osteophytes at baseline and lower osteophyte development at 3years. CONCLUSION Although cigarette smoking did not influence knee function, pain, or the need for replacement surgery, current and former smokers developed fewer osteophytes. This relationship may be linked to the quantity and duration of consumption. Our results provide further insight into the smoking-related pathophysiology of osteoarthritis.
Collapse
Affiliation(s)
- Christian Hubert Roux
- Rheumatology Department, University Nice Sophia Antipolis, LAMHESS, EA 6312, CHU Nice, Nice, France.
| | - Joël Coste
- Biostatistics and Epidemiology Unit, Assistance Publique-Hôpitaux de Paris, Hôtel Dieu, Paris, France
| | - Coralie Roger
- Department of Clinical Research and Innovation, University Hospital of Nice, Nice, France
| | - Eric Fontas
- Department of Clinical Research and Innovation, University Hospital of Nice, Nice, France
| | - Anne-Christine Rat
- CIC 1433 Clinical Epidemiology, Inserm, CHRU University of Lorraine, Nancy, France
| | - Francis Guillemin
- CIC 1433 Clinical Epidemiology, Inserm, CHRU University of Lorraine, Nancy, France
| |
Collapse
|
21
|
Szwedowski D, Szczepanek J, Paczesny Ł, Pękała P, Zabrzyński J, Kruczyński J. Genetics in Cartilage Lesions: Basic Science and Therapy Approaches. Int J Mol Sci 2020; 21:E5430. [PMID: 32751537 PMCID: PMC7432875 DOI: 10.3390/ijms21155430] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/31/2022] Open
Abstract
Cartilage lesions have a multifactorial nature, and genetic factors are their strongest determinants. As biochemical and genetic studies have dramatically progressed over the past decade, the molecular basis of cartilage pathologies has become clearer. Several homeostasis abnormalities within cartilaginous tissue have been found, including various structural changes, differential gene expression patterns, as well as altered epigenetic regulation. However, the efficient treatment of cartilage pathologies represents a substantial challenge. Understanding the complex genetic background pertaining to cartilage pathologies is useful primarily in the context of seeking new pathways leading to disease progression as well as in developing new targeted therapies. A technology utilizing gene transfer to deliver therapeutic genes to the site of injury is quickly becoming an emerging approach in cartilage renewal. The goal of this work is to provide an overview of the genetic basis of chondral lesions and the different approaches of the most recent systems exploiting therapeutic gene transfer in cartilage repair. The integration of tissue engineering with viral gene vectors is a novel and active area of research. However, despite promising preclinical data, this therapeutic concept needs to be supported by the growing body of clinical trials.
Collapse
Affiliation(s)
- Dawid Szwedowski
- Orthopedic Arthroscopic Surgery International (O.A.S.I.) Bioresearch Foundation, Gobbi N.P.O., 20133 Milan, Italy;
- Department of Orthopaedics and Trauma Surgery, Provincial Polyclinical Hospital, 87100 Torun, Poland
| | - Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87100 Torun, Poland
| | - Łukasz Paczesny
- Orvit Clinic, Citomed Healthcare Center, 87100 Torun, Poland; (Ł.P.); (J.Z.)
| | - Przemysław Pękała
- Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, 30705 Krakow, Poland;
| | - Jan Zabrzyński
- Orvit Clinic, Citomed Healthcare Center, 87100 Torun, Poland; (Ł.P.); (J.Z.)
| | - Jacek Kruczyński
- Department of General Orthopaedics, Musculoskeletal Oncology and Trauma Surgery, Poznan University of Medical Sciences, 60512 Poznań, Poland;
| |
Collapse
|
22
|
Łęgosz P, Sarzyńska S, Pulik Ł, Kotrych D, Małdyk P. The complexity of molecular processes in osteoarthritis of the knee joint. Open Med (Wars) 2020; 15:366-375. [PMID: 33335997 PMCID: PMC7711860 DOI: 10.1515/med-2020-0402] [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: 03/27/2019] [Revised: 02/16/2020] [Accepted: 02/25/2020] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is a common medical problem leading to chronic pain and physical disability among the world's population. Analyzing the molecular background of the degenerative arthritis creates the potential for developing novel targeted methods of treatment. Fifty samples of meniscus, anterior cruciate ligaments (ACLs) and articular surfaces were collected from patients who underwent total knee arthroplasty in 2016. Enzyme-linked immunosorbent assay was used to assess the levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF), transforming growth factor-β1 and LUMINEX for MMP-1, MMP-2, MMP-3, MMP-9 and MMP-13. The collected data were correlated with the severity of radiological OA, demographic data and clinical scales. Strong positive correlations in the concentration of metalloproteinases and proinflammatory cytokines, TNF-α (MMP-2 and MMP-13) and IL-6 (MMP-13), were identified. MMP-13 had a positive correlation with the concentration of MMP-1, MMP-2 and MMP-9. Negative correlation coefficient exists between clinical conditions measured with the Western Ontario and McMaster Universities Osteoarthritis Index scale and the level of TNF-α and MMP-1. The TNF-α concentration was lower in the cartilage of the articular surface among patients who took non-steroidal anti-inflammatory drugs periodically. The decrease in MMP-2 in the cartilage of the articular surface corresponded with the severity of radiological OA on the Kellgren-Lawrence scale. Current treatment methods for OA do not stop disease progression. Identifying signaling pathways and molecular particles engaged in OA and their correlations with the patient's clinical condition brings new therapeutic possibilities.
Collapse
Affiliation(s)
- Paweł Łęgosz
- Department of Orthopaedics and Traumatology,
1st Faculty of Medicine, Medical
University of Warsaw, Warsaw, Poland
| | - Sylwia Sarzyńska
- Department of Orthopaedics and Traumatology,
1st Faculty of Medicine, Medical
University of Warsaw, Warsaw, Poland
| | - Łukasz Pulik
- Department of Orthopaedics and Traumatology,
1st Faculty of Medicine, Medical
University of Warsaw, Warsaw, Poland
| | - Daniel Kotrych
- Department of Orthopaedics, Traumatology and
Orthopaedic Oncology, Pomeranian Medical University in
Szczecin, Szczecin, Poland
| | - Paweł Małdyk
- Department of Orthopaedics and Traumatology,
1st Faculty of Medicine, Medical
University of Warsaw, Warsaw, Poland
| |
Collapse
|
23
|
Chien SY, Tsai CH, Liu SC, Huang CC, Lin TH, Yang YZ, Tang CH. Noggin Inhibits IL-1β and BMP-2 Expression, and Attenuates Cartilage Degeneration and Subchondral Bone Destruction in Experimental Osteoarthritis. Cells 2020; 9:cells9040927. [PMID: 32290085 PMCID: PMC7226847 DOI: 10.3390/cells9040927] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a chronic inflammatory and progressive joint disease that results in cartilage degradation and subchondral bone remodeling. The proinflammatory cytokine interleukin 1 beta (IL-1β) is abundantly expressed in OA and plays a crucial role in cartilage remodeling, although its role in the activity of chondrocytes in cartilage and subchondral remodeling remains unclear. In this study, stimulating chondrogenic ATDC5 cells with IL-1β increased the levels of bone morphogenetic protein 2 (BMP-2), promoted articular cartilage degradation, and enhanced structural remodeling. Immunohistochemistry staining and microcomputed tomography imaging of the subchondral trabecular bone region in the experimental OA rat model revealed that the OA disease promotes levels of IL-1β, BMP-2, and matrix metalloproteinase 13 (MMP-13) expression in the articular cartilage and enhances subchondral bone remodeling. The intra-articular injection of Noggin protein (a BMP-2 inhibitor) attenuated subchondral bone remodeling and disease progression in OA rats. We also found that IL-1β increased BMP-2 expression by activating the mitogen-activated protein kinase (MEK), extracellular signal-regulated kinase (ERK), and specificity protein 1 (Sp1) signaling pathways. We conclude that IL-1β promotes BMP-2 expression in chondrocytes via the MEK/ERK/Sp1 signaling pathways. The administration of Noggin protein reduces the expression of IL-1β and BMP-2, which prevents cartilage degeneration and OA development.
Collapse
Affiliation(s)
- Szu-Yu Chien
- Department of Exercise Health Science, National Taiwan University of Sport, Taichung 404393, Taiwan;
- School of Medicine, China Medical University, Taichung 404022, Taiwan;
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 404022, Taiwan;
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung 404022, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin 651012, Taiwan;
| | - Chien-Chung Huang
- School of Medicine, China Medical University, Taichung 404022, Taiwan;
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung 404022, Taiwan
| | - Tzu-Hung Lin
- Material and Chemical Research Laboratories, Industrial Technology Research Institute, Hsinchu 310401, Taiwan; (T.-H.L.); (Y.-Z.Y.)
| | - Yu-Zhen Yang
- Material and Chemical Research Laboratories, Industrial Technology Research Institute, Hsinchu 310401, Taiwan; (T.-H.L.); (Y.-Z.Y.)
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung 404022, Taiwan;
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404022, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 404022, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung 404, Taiwan
- Correspondence: ; Tel.: +886-4-2205-2121 (ext. 7726)
| |
Collapse
|
24
|
Allas L, Rochoux Q, Leclercq S, Boumédiene K, Baugé C. Development of a simple osteoarthritis model useful to predict in vitro the anti-hypertrophic action of drugs. J Transl Med 2020; 100:64-71. [PMID: 31409892 DOI: 10.1038/s41374-019-0303-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/18/2019] [Accepted: 07/07/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is characterized by cartilage degradation, inflammation, and hypertrophy. Therapies are mainly symptomatic and aim to manage pain. Consequently, medical community is waiting for new treatments able to reduce OA process. This study aims to develop an in vitro simple OA model useful to predict drug ability to reduce cartilage hypertrophy. Human primary OA chondrocytes were incubated with transforming growth factor beta 1 (TGF-β1). Hypertrophy was evaluated by Runx2, type X collagen, MMP13, and VEGF expression. Cartilage anabolism was investigated by Sox9, aggrecan, type II collagen, and glycosaminoglycan expression. In chondrocytes, TGF-β1 increased expression of hypertrophic genes and activated canonical WNT pathway, while it decreased dramatically cartilage anabolism, suggesting that this treatment could mimic some OA features in vitro. Additionally, EZH2 inhibition, that has been previously reported to decrease cartilage hypertrophy and reduce OA development in vivo, attenuated COL10A1 and MMP13 upregulation and SOX9 downregulation induced by TGF-β1 treatment. Similarly, pterosin B (an inhibitor of Sik3), and DMOG (a hypoxia-inducible factor prolyl hydroxylase which mimicks hypoxia), repressed the expression of hypertrophy markers in TGF-β stimulated chondrocytes. In conclusion, we established an innovative OA model in vitro. This cheap and simple model will be useful to quickly screen new drugs with potential anti-arthritic effects, in complementary to current inflammatory models, and should permit to accelerate development of efficient treatments against OA able to reduce cartilage hypertrophy.
Collapse
Affiliation(s)
- Lyess Allas
- Normandie Université, UNICAEN, EA7451, BioConnecT, Caen, France
| | - Quitterie Rochoux
- Normandie Université, UNICAEN, EA7451, BioConnecT, Caen, France.,CHU, Service de Rhumatologie, Caen, France
| | - Sylvain Leclercq
- Normandie Université, UNICAEN, EA7451, BioConnecT, Caen, France.,Clinique Saint-Martin, Service de Chirurgie Orthopédique, Caen, France
| | | | - Catherine Baugé
- Normandie Université, UNICAEN, EA7451, BioConnecT, Caen, France.
| |
Collapse
|
25
|
Fibroblast-like synovial cell production of extra domain A fibronectin associates with inflammation in osteoarthritis. BMC Rheumatol 2019; 3:46. [PMID: 31819923 PMCID: PMC6886182 DOI: 10.1186/s41927-019-0093-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
Background The pathophysiology of osteoarthritis (OA) involves wear and tear, and a state of low-grade inflammation. Tissue repair responses include transforming growth factor beta (TGFβ)-induced myofibroblast production of extracellular matrix. Fibronectins are an essential part of the extracellular matrix, and injection of fibronectin fragments into rabbit joints is a previously established animal model of OA. Fibronectin containing the ED-A domain is currently being used as drug delivery target in the development of anti-inflammatory drugs (e.g. Dekavil). Methods In this study, samples of synovial membrane were obtained from patients with knee OA undergoing joint replacement surgery. Immunostaining for ED-A fibronectin and the myofibroblast marker alpha smooth muscle actin (αSMA) was performed on fibroblast-like synovial cells (FLS) and synovial membranes. RAW 264.7 macrophages were incubated with recombinant ED-A fibronectin. Results The staining of ED-A fibronectin in OA FLS was increased by TGFβ but not by TNFα, lipopolysaccharide, or IL-6 (n = 3). ED-A fibronectin co-stained with the myofibroblast marker αSMA in both the OA FLS (n = 3) and in the OA synovial membranes (n = 8). ED-A fibronectin staining was associated with both number of lining layer cells (rho = 0.85 and p = 0.011) and sublining cells (rho = 0.88 and p = 0.007) in the OA synovium (n = 8), and co-distributed with TNFα (n = 5). Recombinant ED-A fibronectin increased the production of TNFα by RAW 264.7 macrophages (n = 3). Conclusions The disease process in OA shares features with the chronic wound healing response. Our findings support utilizing ED-A fibronectin for drug delivery or therapeutic targeting to reduce pro-inflammatory responses in OA.
Collapse
|
26
|
Ahmad M, Hachemi Y, Paxian K, Mengele F, Koenen M, Tuckermann J. A Jack of All Trades: Impact of Glucocorticoids on Cellular Cross-Talk in Osteoimmunology. Front Immunol 2019; 10:2460. [PMID: 31681333 PMCID: PMC6811614 DOI: 10.3389/fimmu.2019.02460] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022] Open
Abstract
Glucocorticoids (GCs) are known to have a strong impact on the immune system, metabolism, and bone homeostasis. While these functions have been long investigated separately in immunology, metabolism, or bone biology, the understanding of how GCs regulate the cellular cross-talk between innate immune cells, mesenchymal cells, and other stromal cells has been garnering attention rather recently. Here we review the recent findings of GC action in osteoporosis, inflammatory bone diseases (rheumatoid and osteoarthritis), and bone regeneration during fracture healing. We focus on studies of pre-clinical animal models that enable dissecting the role of GC actions in innate immune cells, stromal cells, and bone cells using conditional and function-selective mutant mice of the GC receptor (GR), or mice with impaired GC signaling. Importantly, GCs do not only directly affect cellular functions, but also influence the cross-talk between mesenchymal and immune cells, contributing to both beneficial and adverse effects of GCs. Given the importance of endogenous GCs as stress hormones and the wide prescription of pharmaceutical GCs, an improved understanding of GC action is decisive for tackling inflammatory bone diseases, osteoporosis, and aging.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Yasmine Hachemi
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Kevin Paxian
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Florian Mengele
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
- Praxisklinik für Orthopädie, Unfall- und Neurochirurgie Prof. Bischoff/ Dr. Spies/ Dr. Mengele, Neu-Ulm, Germany
| | - Mascha Koenen
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| |
Collapse
|
27
|
|
28
|
Contribution of Infrapatellar Fat Pad and Synovial Membrane to Knee Osteoarthritis Pain. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6390182. [PMID: 31049352 PMCID: PMC6462341 DOI: 10.1155/2019/6390182] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is the most common form of joint disease and a major cause of pain and disability in the adult population. Interestingly, there are patients with symptomatic OA displaying pain, while patients with asymptomatic OA that do not experience pain but show radiographic signs of joint damage. Pain is a complex experience integrating sensory, affective, and cognitive processes related to several peripheral and central nociceptive factors besides inflammation. During the last years, the role of infrapatellar fat pad (IFP), other than the synovial membrane, has been investigated as a potential source of pain in OA. Interestingly, new findings suggest that IFP and synovial membrane might act as a functional unit in OA pathogenesis and pain. The present review discuss the role of IFP and synovial membrane in the development of OA, with a particular focus on pain onset and the possible involved mediators that may play a role in OA pathology and pain mechanisms. Inflammation of IFP and synovial membrane may drive peripheral and central sensitization in KOA. Since sensitization is associated with pain severity in knee OA and may potentially contribute to the transition from acute to chronic, persistent pain in knee OA, preventing sensitization would be a potentially effective and novel means of preventing worsening of pain in knee OA.
Collapse
|
29
|
Park CY. Vitamin D in the Prevention and Treatment of Osteoarthritis: From Clinical Interventions to Cellular Evidence. Nutrients 2019; 11:E243. [PMID: 30678273 PMCID: PMC6413222 DOI: 10.3390/nu11020243] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/03/2019] [Accepted: 01/06/2019] [Indexed: 12/11/2022] Open
Abstract
Older adults are recommended vitamin D to prevent fractures. Though this population is also at risk of osteoarthritis (OA), the effect of vitamin D on OA is unclear and may differ by disease state. The relationship between vitamin D and OA during OA initiation and progression were considered in this narrative review of in vivo and in vitro studies. Regarding OA initiation in humans, the small number of published observational studies suggest a lack of association between induction of OA and vitamin D status. Most randomized controlled trials were performed in White OA patients with relatively high vitamin D status (>50 nmol/L). These studies found no benefit of vitamin D supplementation on OA progression. However, subset analyses and one randomized controlled pilot trial indicated that vitamin D supplementation may alleviate joint pain in OA patients with low vitamin D status (<50 nmol/L). As the etiology of OA is recently being more fully uncovered, better animal and cell models are needed. According to currently available clinical results, evidence is lacking to set a vitamin D level to prevent OA, and increasing vitamin D status above 50 nmol/L does not seem to benefit OA patients.
Collapse
|
30
|
Abstract
Osteoarthritis (OA) is one of the most common diseases, affecting more than 10% of populations and thus creating immense socioeconomic burden. The pathological changes of OA involve the entire joint, which is composed of multiple types of tissues and cells, exemplified by cartilage degradation, subchondral bone thickening, osteophyte formation, synovium inflammation and hypertrophy, and ligament degeneration. As joint homeostasis requires a complex network of growth factors to regulate anabolic and catabolic events, the dysregulation of growth factor signalling would have negative impacts on structure and function of multiple joint tissues and eventually lead to the onset and progression of OA. In this review, we will discuss TGF-β, NGF, Hedgehog and Wnt, the four growth factors which have received extensive attention in the field of OA and clinical/translational interrogation about their application in OA therapies.
Collapse
Affiliation(s)
- Jian Huang
- a Department of Orthopedic Surgery , Rush University Medical Center , Chicago , IL , USA
| | - Lan Zhao
- a Department of Orthopedic Surgery , Rush University Medical Center , Chicago , IL , USA
| | - Di Chen
- a Department of Orthopedic Surgery , Rush University Medical Center , Chicago , IL , USA
| |
Collapse
|
31
|
van der Kraan PM. Differential Role of Transforming Growth Factor-beta in an Osteoarthritic or a Healthy Joint. J Bone Metab 2018; 25:65-72. [PMID: 29900155 PMCID: PMC5995759 DOI: 10.11005/jbm.2018.25.2.65] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/26/2018] [Indexed: 12/24/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine that plays an important role in both normal joints and joints affected by osteoarthritis (OA), the most common joint disease. However, the role of this pleiotropic cytokine in a normal healthy joint is very different from its role in an OA joint. In a normal synovial joint, active TGF-β is only present after joint loading and only for a short period. In contrast, permanent and high levels of active TGF-β are detected in OA joints. Due to this difference in levels and exposure period of joint cells to active TGF-β, the function of TGF-β is strikingly different in normal and OA joints. The consequences of this difference in TGF-β levels on joint homeostasis and pathological changes in OA joints are discussed in this review.
Collapse
Affiliation(s)
- Peter M. van der Kraan
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
32
|
Varela-Eirin M, Loureiro J, Fonseca E, Corrochano S, Caeiro JR, Collado M, Mayan MD. Cartilage regeneration and ageing: Targeting cellular plasticity in osteoarthritis. Ageing Res Rev 2018; 42:56-71. [PMID: 29258883 DOI: 10.1016/j.arr.2017.12.006] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/20/2017] [Accepted: 12/15/2017] [Indexed: 01/15/2023]
Abstract
Ageing processes play a major contributing role for the development of Osteoarthritis (OA). This prototypic degenerative condition of ageing is the most common form of arthritis and is accompanied by a general decline, chronic pain and mobility deficits. The disease is primarily characterized by articular cartilage degradation, followed by subchondral bone thickening, osteophyte formation, synovial inflammation and joint degeneration. In the early stages, osteoarthritic chondrocytes undergo phenotypic changes that increase cell proliferation and cluster formation and enhance the production of matrix-remodelling enzymes. In fact, chondrocytes exhibit differentiation plasticity and undergo phenotypic changes during the healing process. Current studies are focusing on unravelling whether OA is a consequence of an abnormal wound healing response. Recent investigations suggest that alterations in different proteins, such as TGF-ß/BMPs, NF-Kß, Wnt, and Cx43, or SASP factors involved in signalling pathways in wound healing response, could be directly implicated in the initiation of OA. Several findings suggest that osteoarthritic chondrocytes remain in an immature state expressing stemness-associated cell surface markers. In fact, the efficacy of new disease-modifying OA drugs that promote chondrogenic differentiation in animal models indicates that this may be a drug-sensible state. In this review, we highlight the current knowledge regarding cellular plasticity in chondrocytes and OA. A better comprehension of the mechanisms involved in these processes may enable us to understand the molecular pathways that promote abnormal repair and cartilage degradation in OA. This understanding would be advantageous in identifying novel targets and designing therapies to promote effective cartilage repair and successful joint ageing by preventing functional limitations and disability.
Collapse
Affiliation(s)
- Marta Varela-Eirin
- CellCOM research group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | - Jesus Loureiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Eduardo Fonseca
- CellCOM research group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | | | - Jose R Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Manuel Collado
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), SERGAS, Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Maria D Mayan
- CellCOM research group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), Xubias de Arriba, 84, 15006 A Coruña, Spain.
| |
Collapse
|
33
|
The Clinical Significance of Osteophytes in Compartments of the Knee Joint With Normal Articular Cartilage. AJR Am J Roentgenol 2018; 210:W164-W171. [PMID: 29470158 DOI: 10.2214/ajr.17.18664] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The purpose of this study is to determine whether marginal osteophytes in compartments with normal cartilage would be more frequently observed in knees with cartilage lesions and osteophytes in other compartments. MATERIALS AND METHODS This retrospective study reviewed 500 consecutive knee MRI examinations performed within 6 months of arthroscopic knee surgery conducted for 497 patients with symptoms (289 male patients and 208 female patients; age range, 17-74 years; median age, 43 years). The highest grade of cartilage lesion detected at MRI and arthroscopy was recorded. Marginal osteophytes were graded on MRI with use of a standardized scoring system, with grade 0 denoting no osteophyte; grade 1, small osteophyte; grade 2, medium-size osteophyte; and grade 3, large osteophyte). The frequency of false-positive osteophytes, defined as osteophytes present in compartments (the patellofemoral, medial tibiofemoral, and lateral tibiofemoral compartments) with normal cartilage observed on MRI and arthroscopy, was calculated. The Goodman and Kruskal gamma statistic was used to test the association of osteophyte size between compartments. Logistic regression was used to test the association between osteophyte size and the severity of the cartilage lesions. RESULTS Marginal osteophytes were seen in compartments with normal cartilage on MRI and arthroscopy in 60.5% of knees (75 of 124) with cartilage lesions and osteophytes in other compartments and accounted for all false-positive grade 2 and grade 3 osteophytes. Marginal osteophytes were seen in 12.7% of knees (13 of 102) that had no cartilage lesions in any compartment on MRI or arthroscopy, and all of these were grade 1 osteophytes. The presence of larger sized osteophytes in the compartments with cartilage lesions was associated with the presence of larger sized osteophytes in the compartments with normal cartilage. More severe cartilage lesions were associated with larger osteophyte size. CONCLUSION Compartments with marginal osteophytes and normal cartilage are commonly seen in knees that have other compartments with osteophytes and cartilage lesions.
Collapse
|
34
|
Stem Cells for Cartilage Repair: Preclinical Studies and Insights in Translational Animal Models and Outcome Measures. Stem Cells Int 2018. [PMID: 29535784 PMCID: PMC5832141 DOI: 10.1155/2018/9079538] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Due to the restricted intrinsic capacity of resident chondrocytes to regenerate the lost cartilage postinjury, stem cell-based therapies have been proposed as a novel therapeutic approach for cartilage repair. Moreover, stem cell-based therapies using mesenchymal stem cells (MSCs) or induced pluripotent stem cells (iPSCs) have been used successfully in preclinical and clinical settings. Despite these promising reports, the exact mechanisms underlying stem cell-mediated cartilage repair remain uncertain. Stem cells can contribute to cartilage repair via chondrogenic differentiation, via immunomodulation, or by the production of paracrine factors and extracellular vesicles. But before novel cell-based therapies for cartilage repair can be introduced into the clinic, rigorous testing in preclinical animal models is required. Preclinical models used in regenerative cartilage studies include murine, lapine, caprine, ovine, porcine, canine, and equine models, each associated with its specific advantages and limitations. This review presents a summary of recent in vitro data and from in vivo preclinical studies justifying the use of MSCs and iPSCs in cartilage tissue engineering. Moreover, the advantages and disadvantages of utilizing small and large animals will be discussed, while also describing suitable outcome measures for evaluating cartilage repair.
Collapse
|
35
|
Hsia AW, Emami AJ, Tarke FD, Cunningham HC, Tjandra PM, Wong A, Christiansen BA, Collette NM. Osteophytes and fracture calluses share developmental milestones and are diminished by unloading. J Orthop Res 2018; 36:699-710. [PMID: 29058776 PMCID: PMC5877458 DOI: 10.1002/jor.23779] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/13/2017] [Indexed: 02/04/2023]
Abstract
Osteophytes are a typical radiographic finding during osteoarthritis (OA), but the mechanisms leading to their formation are not well known. Comparatively, fracture calluses have been studied extensively; therefore, drawing comparisons between osteophytes and fracture calluses may lead to a deeper understanding of osteophyte formation. In this study, we compared the time courses of osteophyte and fracture callus formation, and investigated mechanisms contributing to development of these structure. Additionally, we investigated the effect of mechanical unloading on the formation of both fracture calluses and osteophytes. Mice underwent either transverse femoral fracture or non-invasive anterior cruciate ligament rupture. Fracture callus and osteophyte size and ossification were evaluated after 3, 5, 7, 14, 21, or 28 days. Additional mice were subjected to hindlimb unloading after injury for 3, 7, or 14 days. Protease activity and gene expression profiles after injury were evaluated after 3 or 7 days of normal ambulation or hindlimb unloading using in vivo fluorescence reflectance imaging (FRI) and quantitative PCR. We found that fracture callus and osteophyte growth achieved similar developmental milestones, but fracture calluses formed and ossified at earlier time points. Hindlimb unloading ultimately led to a threefold decrease in chondro/osteophyte area, and a twofold decrease in fracture callus area. Unloading was also associated with decreased inflammation and protease activity in injured limbs detected with FRI, particularly following ACL rupture. qPCR analysis revealed disparate cellular responses in fractured femurs and injured joints, suggesting that fracture calluses and osteophytes may form via different inflammatory, anabolic, and catabolic pathways. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:699-710, 2018.
Collapse
Affiliation(s)
- Allison W. Hsia
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
| | - Armaun J. Emami
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
| | - Franklin D. Tarke
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA
| | - Hailey C. Cunningham
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
| | - Priscilla M. Tjandra
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
| | - Alice Wong
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA
| | - Blaine A. Christiansen
- Biomedical Engineering Graduate Group, University of California Davis, Davis, CA
- Department of Orthopaedic Surgery, University of California Davis Medical Center, Sacramento, CA
| | - Nicole M. Collette
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA
| |
Collapse
|
36
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
Abstract
TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
37
|
Modulation of TGF‑β activity by latent TGF‑β‑binding protein 1 in human osteoarthritis fibroblast‑like synoviocytes. Mol Med Rep 2017; 17:1893-1900. [PMID: 29257223 DOI: 10.3892/mmr.2017.8086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 10/20/2017] [Indexed: 11/05/2022] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease; however, its underlying pathogenesis remains to be elucidated. Previous studies have demonstrated that the transforming growth factor‑β (TGF‑β) signaling pathway has a role in the initiation and development of OA. Additionally, latent TGF‑β‑binding protein‑1 (LTBP‑1) modulates the activity of the TGF‑β‑mothers against decapentaplegic (Smad) signaling pathway in numerous diseases, including malignant glioma. The present study demonstrated that expression of LTBP‑1 is increased in OA synovial tissues compared with normal synovial tissues. The effect of TGF‑β was identified to be mediated by phosphorylated(p)‑(Smad)2/3, which may activate activin‑like kinase (ALK)5 receptor, and by p‑Smad1/5/8, which may induce ALK1, thereby stimulating expression of matrix metalloproteinase‑(MMP)‑13 in OA fibroblast‑like synoviocytes (FLS). Compared with normal FLS, OA FLS demonstrated an increased p‑Smad1/5/8:p‑Smad2 ratio, which led to elevated MMP‑13 expression and aggravation of OA. Furthermore, knockdown of the LTBP‑1 gene by siRNA transfection in OA FLS reduced p‑Smad1/5/8 expression without affecting TGF‑β mRNA levels, although p‑Smad2 expression increased. It was also demonstrated that OA FLS exhibited increased proliferation compared with normal FLS in vitro. Furthermore, siRNA‑mediated downregulation of LTBP‑1 reduced proliferation of OA FLS. In conclusion, the present study demonstrated that an alteration in the p‑Smad1/5/8:p‑Smad2 ratio as well as association between p‑Smad1/5/8 and MMP‑13 expression in human OA FLS, may contribute to the development of OA. The results of the present study suggested that LTBP‑1 is a modulator of the TGF‑β signaling pathway in human OA FLS, which may aid in elucidating the mechanism underlying the pathology of OA.
Collapse
|
38
|
Crema MD, Felson DT, Guermazi A, Nevitt MC, Niu J, Lynch JA, Marra MD, Torner J, Lewis CE, Roemer FW. Is the atrophic phenotype of tibiofemoral osteoarthritis associated with faster progression of disease? The MOST study. Osteoarthritis Cartilage 2017; 25:1647-1653. [PMID: 28606556 PMCID: PMC5605441 DOI: 10.1016/j.joca.2017.05.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 03/31/2017] [Accepted: 05/31/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To assess the associations of atrophic tibiofemoral osteoarthritis (OA) with progression of radiographic joint space narrowing (JSN) and magnetic resonance imaging (MRI)-defined progression of cartilage damage. DESIGN Participants of the Multicenter Osteoarthritis (MOST) Study with available radiographic and MRI assessments at baseline and 30 months were included. The atrophic OA phenotype was defined as Osteoarthritis Research Society International (OARSI) grades 1 or 2 for JSN and grade 0 for osteophytes. Based on MRI, atrophic OA was defined as tibiofemoral (TF) cartilage damage grades ≥3 in at least 2 of 10 subregions with absent or tiny osteophytes in all TF subregions. Progression of JSN and cartilage loss on MRI, was defined as (1) no, (2) slow, and (3) fast progression. Co-variance and logistic regression with generalized estimated equations were performed to assess the association of atrophic knee OA with any progression, compared to non-atrophic OA knees. RESULTS A total of 476 knees from 432 participants were included. There were 50 (10.5%) knees with atrophic OA using the radiographic definition, and 16 (3.4%) knees with atrophic OA using MRI definition. Non-atrophic OA knees more commonly exhibited fast progression of JSN and cartilage damage. Logistic regression showed that the atrophic phenotype of knee OA was associated with a decreased likelihood of progression of JSN and cartilage loss. CONCLUSION In this sample, the atrophic phenotype of knee OA was associated with a decreased likelihood of progression of JSN and cartilage loss compared to the non-atrophic knee OA phenotype.
Collapse
Affiliation(s)
- M D Crema
- Department of Radiology, Quantitative Imaging Center, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, Saint-Antoine Hospital, University Paris VI, Paris, France.
| | - D T Felson
- Clinical Epidemiology Research and Training Unit, Boston University, Boston, MA, USA
| | - A Guermazi
- Department of Radiology, Quantitative Imaging Center, Boston University School of Medicine, Boston, MA, USA
| | - M C Nevitt
- Department of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, USA
| | - J Niu
- Clinical Epidemiology Research and Training Unit, Boston University, Boston, MA, USA
| | - J A Lynch
- Department of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, USA
| | - M D Marra
- Department of Radiology, Quantitative Imaging Center, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, Saint-Antoine Hospital, University Paris VI, Paris, France
| | - J Torner
- University of Iowa, Iowa City, IA, USA
| | - C E Lewis
- University of Alabama, Birmingham, AL, USA
| | - F W Roemer
- Department of Radiology, Quantitative Imaging Center, Boston University School of Medicine, Boston, MA, USA; Department of Radiology, University of Erlangen, Erlangen, Germany
| |
Collapse
|
39
|
Zhang L, Wang PE, Ying J, Jin X, Luo C, Xu T, Xu S, Dong R, Xiao L, Tong P, Jin H. Yougui Pills Attenuate Cartilage Degeneration via Activation of TGF-β/Smad Signaling in Chondrocyte of Osteoarthritic Mouse Model. Front Pharmacol 2017; 8:611. [PMID: 28928664 PMCID: PMC5591843 DOI: 10.3389/fphar.2017.00611] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Accepted: 08/23/2017] [Indexed: 01/15/2023] Open
Abstract
Yougui pills (YGPs) have been used for centuries in the treatment of Chinese patients with Kidney-Yang Deficiency Syndrome. Despite the fact that the efficiency of YGPs on treating osteoarthritis has been verified in clinic, the underlying mechanisms are not totally understood. The present study observes the therapeutic role of YGPs and mechanisms underlying its chondroprotective action in osteoarthritic cartilage. To evaluate the chondroprotective effects of YGPs, we examined the impact of orally administered YGPs in a model of destabilization of the medial meniscus (DMM). Male C57BL/6J mice were provided a daily treatment of YGPs and a DMM surgery was performed on the right knee. At 12 weeks post-surgery, the joints were harvested for tissue analyses, including histomorphometry, OARSI scoring, micro-CT and immunohistochemistry for COL-2, MMP-13 and pSMAD-2. We also performed the relative experiments mentioned above in mice with Tgfbr2 conditional knockout (TGF-βRIICol2ER mice) in articular cartilage. To evaluate the safety of YGPs, hematology was determined in each group. Amelioration of cartilage degradation was observed in the YGPs group, with increases in cartilage area and thickness, proteoglycan matrix, and decreases in OARSI score at 12 weeks post surgery. In addition, reduced BV/TV and Tb. Th, and elevated Tb. Sp were observed in DMM-induced mice followed by YGPs treatment. Moreover, the preservation of cartilage correlated with reduced MMP-13, and elevated COL-2 and pSMAD-2 protein expressional levels were also revealed in DMM-induced mice treated with YGPs. Similarly, TGF-βRIICol2ER mice exhibited significant OA-like phenotype. However, no significant difference in cartilage structure was observed in TGF-βRIICol2ER mice after YGPs treatment. Interestingly, no obvious adverse effects were observed in mice from each group based on the hematologic analyses. These findings suggested that YGPs could inhibit cartilage degradation through enhancing TGF-β/Smad signaling activation, and be considered a good option for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Lei Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Ping-Er Wang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China
| | - Jun Ying
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Xing Jin
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,Department of Orthopaedics and Traumatology, Wangjiang Sub-District Community Health Service CenterHangzhou, China
| | - Cheng Luo
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Taotao Xu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Shibing Xu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Rui Dong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China.,The First College of Clinical Medicine, Zhejiang Chinese Medical UniversityHangzhou, China
| | - Luwei Xiao
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China
| | - Peijian Tong
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China
| | - Hongting Jin
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, China
| |
Collapse
|
40
|
Hu Y, Yang Y, Luo B. Evaluation of destruction in a collagen-induced arthritis rat model: Bony spur formation. Exp Ther Med 2017; 14:2563-2567. [PMID: 28962195 PMCID: PMC5609218 DOI: 10.3892/etm.2017.4817] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 04/21/2017] [Indexed: 12/18/2022] Open
Abstract
Over the past 40 years, the collagen-induced arthritis (CIA) animal model has been widely used as a model of rheumatoid arthritis (RA). However, no model is able to completely depict the characteristics of cartilage destruction to date. In the later stage of joint cartilage destruction, bony spurs form in RA. This bony spur formation is an important symptom in the pathological development of RA. In the present study, CIA was used to elucidate the pathological process of bony spur formation. Joint damage and spur formation in the animal model was detected by radiology and histology. Radiology identified bony spurs in the knee and foot joints, which worsened as the disease progressed. Furthermore, following observations of histological sections, fusion and damage of the articular cartilage, as well as a higher number of osteoclasts, were identified. Previous results have determined that bony spurs may be involved in another pathological process that occurs during the later stages of RA. Therefore, further studies investigating this symptom are required to improve the understanding of RA and facilitate the development of an appropriate treatment for RA.
Collapse
Affiliation(s)
- Yiping Hu
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, P.R. China.,Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, Guangdong 518055, P.R. China
| | - Yi Yang
- Department of Gynecology, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Bin Luo
- Department of Cardiac and Vascular Surgery, Shenzhen People's Hospital, Jinan University, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
41
|
Hamada D, Maynard R, Schott E, Drinkwater CJ, Ketz JP, Kates SL, Jonason JH, Hilton MJ, Zuscik MJ, Mooney RA. Suppressive Effects of Insulin on Tumor Necrosis Factor-Dependent Early Osteoarthritic Changes Associated With Obesity and Type 2 Diabetes Mellitus. Arthritis Rheumatol 2017; 68:1392-402. [PMID: 26713606 PMCID: PMC4882234 DOI: 10.1002/art.39561] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 12/17/2015] [Indexed: 12/24/2022]
Abstract
Objective Obesity is a state of chronic inflammation that is associated with insulin resistance and type 2 diabetes mellitus (DM), as well as an increased risk of osteoarthritis (OA). This study was undertaken to define the links between obesity‐associated inflammation, insulin resistance, and OA, by testing the hypotheses that 1) tumor necrosis factor (TNF) is critical in mediating these pathologic changes in OA, and 2) insulin has direct effects on the synovial joint that are compromised by insulin resistance. Methods The effects of TNF and insulin on catabolic gene expression were determined in fibroblast‐like synoviocytes (FLS) isolated from human OA synovium. Synovial TNF expression and OA progression were examined in 2 mouse models, high‐fat (HF) diet–fed obese mice with type 2 DM and TNF‐knockout mice. Insulin resistance was investigated in synovium from patients with type 2 DM. Results Insulin receptors (IRs) were abundant in both mouse and human synovial membranes. Human OA FLS were insulin responsive, as indicated by the dose‐dependent phosphorylation of IRs and Akt. In cultures of human OA FLS with exogenous TNF, the expression and release of MMP1, MMP13, and ADAMTS4 by FLS were markedly increased, whereas after treatment with insulin, these effects were selectively inhibited by >50%. The expression of TNF and its abundance in the synovium were elevated in samples from obese mice with type 2 DM. In TNF‐knockout mice, increases in osteophyte formation and synovial hyperplasia associated with the HF diet were blunted. The synovium from OA patients with type 2 DM contained markedly more macrophages and showed elevated TNF levels as compared to the synovium from OA patients without diabetes. Moreover, insulin‐dependent phosphorylation of IRs and Akt was blunted in cultures of OA FLS from patients with type 2 DM. Conclusion TNF appears to be involved in mediating the advanced progression of OA seen in type 2 DM. While insulin plays a protective, antiinflammatory role in the synovium, insulin resistance in patients with type 2 DM may impair this protective effect and promote the progression of OA.
Collapse
Affiliation(s)
- Daisuke Hamada
- University of Rochester Medical Center, Rochester, New York
| | - Robert Maynard
- University of Rochester Medical Center, Rochester, New York
| | - Eric Schott
- University of Rochester Medical Center, Rochester, New York
| | | | - John P Ketz
- University of Rochester Medical Center, Rochester, New York
| | | | | | | | | | | |
Collapse
|
42
|
Kaneguchi A, Ozawa J, Kawamata S, Yamaoka K. Development of arthrogenic joint contracture as a result of pathological changes in remobilized rat knees. J Orthop Res 2017; 35:1414-1423. [PMID: 27601089 DOI: 10.1002/jor.23419] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/31/2016] [Indexed: 02/04/2023]
Abstract
This study aimed to elucidate how rats recover from immobilization-induced knee joint contracture. Rats' right knees were immobilized by an external fixator at a flexion of 140° for 3 weeks. After removal of the fixator, the joints were allowed to move freely (remobilization) for 0, 1, 3, 7, or 14 days (n = 5 each). To distinguish myogenic and arthrogenic contractures, the passive extension range of motion was measured before and after myotomy of the knee flexors. Knee joints were histologically analyzed and the expression of genes encoding inflammatory or fibrosis-related mediators, interleukin-1β (1L-1β), fibrosis-related transforming growth factor-β1 (TGF-β1), and collagen type I (COL1A1) and III (COL3A1), were examined in the knee joint posterior capsules using real-time PCR. Both myogenic and arthrogenic contractures were established within 3 weeks of immobilization. During remobilization, the myogenic contracture decreased over time. In contrast, the arthrogenic contracture developed further during the remobilization period. On day 1 of remobilization, inflammatory changes characterized by edema, inflammatory cell infiltration, and upregulation of IL-1β gene started in the knee joint posterior capsule. In addition, collagen deposition accompanied by fibroblast proliferation, with upregulation of TGF-β1, COL1A1, and COL3A1 genes, appeared in the joint capsule between days 7 and 14. These results suggest the progression of arthrogenic contracture following remobilization, which is characterized by fibrosis development, is possibly triggered by inflammation in the joint capsule. It is therefore necessary to focus on developing new treatment strategies for immobilization-induced joint contracture. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1414-1423, 2017.
Collapse
Affiliation(s)
- Akinori Kaneguchi
- Major in Medical Engineering and Technology, Graduate School of Medical Technology and Health Welfare Sciences, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima, Japan
| | - Junya Ozawa
- Faculty of Rehabilitation, Department of Rehabilitation, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima 739-2695, Japan
| | - Seiichi Kawamata
- Institute of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-Ku, Hiroshima, Japan
| | - Kaoru Yamaoka
- Faculty of Rehabilitation, Department of Rehabilitation, Hiroshima International University, Kurose-Gakuendai 555-36, Higashi-Hiroshima, Hiroshima 739-2695, Japan
| |
Collapse
|
43
|
|
44
|
Changes in the osteochondral unit during osteoarthritis: structure, function and cartilage-bone crosstalk. Nat Rev Rheumatol 2016; 12:632-644. [PMID: 27652499 DOI: 10.1038/nrrheum.2016.148] [Citation(s) in RCA: 521] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In diarthrodial joints, the articular cartilage, calcified cartilage, and subchondral cortical and trabecular bone form a biocomposite - referred to as the osteochondral unit - that is uniquely adapted to the transfer of load. During the evolution of the osteoarthritic process the compositions, functional properties, and structures of these tissues undergo marked alterations. Although pathological processes might selectively target a single joint tissue, ultimately all of the components of the osteochondral unit will be affected because of their intimate association, and thus the biological and physical crosstalk among them is of great importance. The development of targeted therapies against the osteoarthritic processes in cartilage or bone will, therefore, require an understanding of the state of these joint tissues at the time of the intervention. Importantly, these interventions will not be successful unless they are applied at the early stages of disease before considerable structural and functional alterations occur in the osteochondral unit. This Review describes the changes that occur in bone and cartilage during the osteoarthritic process, and highlights strategies for how this knowledge could be applied to develop new therapeutic interventions for osteoarthritis.
Collapse
|
45
|
Hu XK, Yin XH, Zhang HQ, Guo CF, Tang MX. Liraglutide attenuates the osteoblastic differentiation of MC3T3‑E1 cells by modulating AMPK/mTOR signaling. Mol Med Rep 2016; 14:3662-8. [PMID: 27600753 PMCID: PMC5042760 DOI: 10.3892/mmr.2016.5729] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 07/29/2016] [Indexed: 01/13/2023] Open
Abstract
Liraglutide, a synthetic analogue of glucagon-like peptide-1, is utilized in the treatment of type 2 diabetes and obesity. Liraglutide has been previously demonstrated to prevent osteoblastic differentiation of human vascular smooth muscle cells, resulting in the slowing of arterial calcification, however, its effect on bone formation remains unclear. The present study investigated the effect of liraglutide on osteoblastic differentiation using Alizarin Red S staining, and examined the molecular mechanisms underlying the regulatory effect by western blot analysis. The present study demonstrated that protein expression levels of phosphorylated adenosine monophosphate-activated protein kinase (p-AMPK) were downregulated in MC3T3-E1 cells during osteoblastic differentiation in commercial osteogenic differentiation medium, whereas protein expression levels of transforming growth factor-β (TGF-β) and phosphorylated mammalian target of rapamycin (p-mTOR) increased. Liraglutide was subsequently demonstrated to dose-dependently attenuate the osteoblastic differentiation of MC3T3-E1 cells, to upregulate p-AMPK, and downregulate p-mTOR and TGF-β protein expression levels. Treatment with an AMPK-specific inhibitor, Compound C, eradicated the effect of liraglutide on osteoblastic differentiation, and p-mTOR and TGF-β downregulation. An mTOR activator, MHY1485, also abolished the inhibitory effect of liraglutide on osteoblastic differentiation, and resulted in p-mTOR and TGF-β downregulation, but did not attenuate the liraglutide-induced increase in p-AMPK protein expression levels. The results of the present study demonstrate that liraglutide attenuates osteoblastic differentiation of MC3T3-E1 cells via modulation of AMPK/mTOR signaling. The present study revealed a novel function of liraglutide, which contributes to the understanding of its pharmacological and physiological effects in clinical settings.
Collapse
Affiliation(s)
- Xiong-Ke Hu
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Xin-Hua Yin
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hong-Qi Zhang
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Chao-Feng Guo
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Ming-Xing Tang
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
46
|
van der Kraan P, Matta C, Mobasheri A. Age-Related Alterations in Signaling Pathways in Articular Chondrocytes: Implications for the Pathogenesis and Progression of Osteoarthritis - A Mini-Review. Gerontology 2016; 63:29-35. [PMID: 27595269 DOI: 10.1159/000448711] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/27/2016] [Indexed: 11/19/2022] Open
Abstract
Musculoskeletal conditions are a major burden on individuals, healthcare systems, and social care systems throughout the world, with indirect costs having a predominant economic impact. Aging is a major contributing factor to the development and progression of arthritic and musculoskeletal diseases. Indeed, aging and inflammation (often referred to as 'inflammaging') are critical risk factors for the development of osteoarthritis (OA), which is one of the most common forms of joint disease. The term 'chondrosenescence' has recently been introduced to define the age-dependent deterioration of chondrocyte function and how it undermines cartilage function in OA. An important component of chondrosenescence is the age-related deregulation of subcellular signaling pathways in chondrocytes. This mini-review discusses the role of age-related alterations in chondrocyte signaling pathways. We focus our attention on two major areas: age-dependent alterations in transforming growth factor-β signaling and changes in protein kinase and phosphoprotein phosphatase activities in aging chondrocytes. A better understanding of the basic signaling mechanisms underlying aging in chondrocytes is likely to facilitate the development of new therapeutic and preventive strategies for OA and a range of other age-related osteoarticular disorders.
Collapse
Affiliation(s)
- Peter van der Kraan
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | |
Collapse
|
47
|
Kerkhofs J, Leijten J, Bolander J, Luyten FP, Post JN, Geris L. A Qualitative Model of the Differentiation Network in Chondrocyte Maturation: A Holistic View of Chondrocyte Hypertrophy. PLoS One 2016; 11:e0162052. [PMID: 27579819 PMCID: PMC5007039 DOI: 10.1371/journal.pone.0162052] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/18/2016] [Indexed: 01/15/2023] Open
Abstract
Differentiation of chondrocytes towards hypertrophy is a natural process whose control is essential in endochondral bone formation. It is additionally thought to play a role in several pathophysiological processes, with osteoarthritis being a prominent example. We perform a dynamic analysis of a qualitative mathematical model of the regulatory network that directs this phenotypic switch to investigate the influence of the individual factors holistically. To estimate the stability of a SOX9 positive state (associated with resting/proliferation chondrocytes) versus a RUNX2 positive one (associated with hypertrophy) we employ two measures. The robustness of the state in canalisation (size of the attractor basin) is assessed by a Monte Carlo analysis and the sensitivity to perturbations is assessed by a perturbational analysis of the attractor. Through qualitative predictions, these measures allow for an in silico screening of the effect of the modelled factors on chondrocyte maintenance and hypertrophy. We show how discrepancies between experimental data and the model’s results can be resolved by evaluating the dynamic plausibility of alternative network topologies. The findings are further supported by a literature study of proposed therapeutic targets in the case of osteoarthritis.
Collapse
Affiliation(s)
- Johan Kerkhofs
- Biomechanics Research Unit, University of Liège, Liège, Belgium
- Biomechanics section, KU Leuven, Leuven, Belgium
- Prometheus, the Leuven R&D division of skeletal tissue engineering, KU Leuven, Leuven, Belgium
| | - Jeroen Leijten
- Prometheus, the Leuven R&D division of skeletal tissue engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Johanna Bolander
- Prometheus, the Leuven R&D division of skeletal tissue engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Frank P. Luyten
- Prometheus, the Leuven R&D division of skeletal tissue engineering, KU Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Janine N. Post
- Developmental BioEngineering, MIRA Institute for biomedical technology and technical medicine, University of Twente, Enschede, The Netherlands
| | - Liesbet Geris
- Biomechanics Research Unit, University of Liège, Liège, Belgium
- Biomechanics section, KU Leuven, Leuven, Belgium
- Prometheus, the Leuven R&D division of skeletal tissue engineering, KU Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
48
|
Ligustrazine Inhibits Cartilage Endplate Hypertrophy via Suppression of TGF-β1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:1042489. [PMID: 27563332 PMCID: PMC4985580 DOI: 10.1155/2016/1042489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/27/2016] [Indexed: 11/17/2022]
Abstract
CEP hypertrophy is one of the characteristics of intervertebral disc degeneration (IDD). LIG exerts a protective effect on IDD in animal model. The effect of LIG on CEP hypertrophy is further investigated in the present study. Cells were isolated from hypertrophic samples obtained from patients during vertebral fusion surgery. Cellular proliferation and the expression of type I collagen (Col I) and TGF-β1 were tested. In the bipedal rats, the edges of the CEP and the sizes of noncartilaginous outgrowth, as well as the expression of osteogenic markers, Col1a, ALP, Runx2, and TGF-β1, were detected. Within two passages, the condensed hypertrophic CEP cells exhibited osteogenic capacity by bony-like nodules and ALP positive staining, along with increased expression of Col I and TGF-β1. LIG inhibited proliferation of CEP cells and downregulated the expression of Col I and TGF-β1 in vitro. Furthermore, LIG attenuated CEP hypertrophy on the lumbar spine of bipedal rats by reducing Col1a, ALP, Runx2, and TGF-β1 mRNA expression and TGF-β1 distribution in vivo. We concluded LIG exerted a preventive effect on CEP hypertrophy via suppression of TGF-β1 levels. This information could be used to develop alternative therapeutic methods to treat spinal CEP hypertrophy.
Collapse
|
49
|
LIU SHENSHEN, ZHOU PU, ZHANG YANQIU. Abnormal expression of key genes and proteins in the canonical Wnt/β-catenin pathway of articular cartilage in a rat model of exercise-induced osteoarthritis. Mol Med Rep 2016; 13:1999-2006. [PMID: 26794964 PMCID: PMC4768959 DOI: 10.3892/mmr.2016.4798] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 12/23/2015] [Indexed: 01/16/2023] Open
Abstract
To investigate the molecular pathogenesis of the canonical Wnt/β-catenin pathway in exercise-induced osteoarthritis (OA), 30 male healthy Sprague Dawley rats were divided into three groups (control, normal exercise‑induced OA and injured exercise‑induced OA groups) in order to establish the exercise‑induced OA rat model. The mRNA and protein expression levels of Runx‑2, BMP‑2, Ctnnb1, Sox‑9, collagen Ⅱ, Mmp‑13, Wnt‑3a and β‑catenin in chondrocytes were detected by reverse transcription‑quantitative polymerase chain reaction, western blotting and immunohistochemical staining. The mRNA levels of Runx‑2, BMP‑2 and Ctnnb1 were upregulated in the normal exercise‑induced OA and injured exercise‑induced OA groups; while Runx‑2 and BMP‑2 were upregulated in the injured exercise‑induced OA group when compared with the normal exercise‑induced OA group. The protein levels of Mmp‑13, Wnt‑3a and β‑catenin were increased and collagen Ⅱ was reduced in the normal exercise‑induced OA and injured exercise‑induced OA groups. Ctnnb1, Wnt‑3a and β‑catenin, which are key genes and proteins in the canonical Wnt/β‑catenin pathway, were abnormally expressed in chondrocytes of the exercise‑induced OA rat model. Ctnnb1, β‑catenin and Wnt‑3a were suggested to participate in the pathogenesis of exercise‑induced OA by abnormally activating the Wnt/β‑catenin pathway during physical exercise due to excessive pressure. The results of the present study may provide an improved understanding of the pathogenesis of exercise-induced OA.
Collapse
Affiliation(s)
- SHEN-SHEN LIU
- College of Physical Education, Langfang Teachers University, Langfang, Hebei 065000, P.R. China
| | - PU ZHOU
- College of Physical Education, Langfang Teachers University, Langfang, Hebei 065000, P.R. China
| | - YANQIU ZHANG
- Department of Physical Education, Xi'an Shiyou University, Xi'an, Shaanxi 710065, P.R. China
| |
Collapse
|
50
|
Mechanobiology of TGFβ signaling in the skeleton. Matrix Biol 2016; 52-54:413-425. [PMID: 26877077 DOI: 10.1016/j.matbio.2016.02.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/05/2016] [Accepted: 02/08/2016] [Indexed: 12/12/2022]
Abstract
Physical and biochemical cues play fundamental roles in the skeleton at both the tissue and cellular levels. The precise coordination of these cues is essential for skeletal development and homeostasis, and disruption of this coordination can drive disease progression. The growth factor TGFβ is involved in both the regulation of and cellular response to the physical microenvironment. It is essential to summarize the current findings regarding the mechanisms by which skeletal cells integrate physical and biochemical cues so that we can identify and address remaining gaps that could ultimately improve skeletal health. In this review, we describe the role of TGFβ in mechanobiological signaling in bone and cartilage at the tissue and cellular levels. We provide detail on how static and dynamic physical cues at the macro-level are transmitted to the micro-level, ultimately leading to regulation at each level of the TGFβ pathway and to cell differentiation. The continued integration of engineering and biological approaches is needed to answer many remaining questions, such as the mechanisms by which cells generate a coordinated response to physical and biochemical cues. We propose one such mechanism, through which the combination of TGFβ and an optimal physical microenvironment leads to synergistic induction of downstream TGFβ signaling.
Collapse
|