1
|
Lee YM, Son E, Kim DS, Shim KS, Yu SH. Evaluating the Anti-Inflammatory and Chondroprotective Effects of Adenocaulon himalaicum Extract Through Network Pharmacology and Experimental Validation. Int J Mol Sci 2025; 26:877. [PMID: 39940649 PMCID: PMC11816759 DOI: 10.3390/ijms26030877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/23/2024] [Accepted: 01/16/2025] [Indexed: 02/16/2025] Open
Abstract
Conventional osteoarthritis treatments have several side effects and poor efficacy. This study explored the anti-inflammatory and cartilage-protective effects of Adenocaulon himalaicum, with a focus on its potential application in osteoarthritis treatment. The anti-inflammatory effects of A. himalaicum extract (AHLE) were investigated in lipopolysaccharide-induced RAW264.7 macrophages, interleukin (IL)-1β-stimulated chondrocytes, and rats with carrageenan-induced hind paw oedema. We also evaluated AHLE's analgesic activity in mice with acetic acid-induced writhing. The components of AHLE were subjected to network pharmacological analysis to elucidate their mechanisms of action and validate potential pathways and targets in vitro. AHLE markedly reduced nitric oxide, IL-1β, IL-6, tumour necrosis factor-alpha, and prostaglandin E2 production in both RAW264.7 macrophages and chondrocytes. In animal models, AHLE reduced carrageenan-induced hind paw swelling and provided analgesic effects in writhing tests. The main components were chlorogenic acid; 1,3-dicaffeoylquinic acid; 3,4-dicaffeoylquinic acid; 3,5-dicaffeoylquinic acid; and 4,5-dicaffeoylquinic acid. According to network pharmacological analysis, AHLE's main therapeutic targets are the mitogen-activated protein kinase (MAPK) signalling pathway and extracellular matrix (ECM) degradation. These targets were verified through the MAPK pathway and expression of matrix metalloproteinase, an enzyme involved in ECM degradation. In conclusion, AHLE has considerable anti-inflammatory and cartilage-protective properties, making it a promising candidate for osteoarthritis therapy.
Collapse
Affiliation(s)
- Yun Mi Lee
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Eunjung Son
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Dong-Seon Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Kyu-Suk Shim
- Univera Co., Ltd., Cheonan 31257, Republic of Korea
| | - Su Hyun Yu
- Univera Co., Ltd., Cheonan 31257, Republic of Korea
| |
Collapse
|
2
|
Zhuang H, Ren X, Zhang Y, Li H, Zhou P. β-Hydroxybutyrate enhances chondrocyte mitophagy and reduces cartilage degeneration in osteoarthritis via the HCAR2/AMPK/PINK1/Parkin pathway. Aging Cell 2024; 23:e14294. [PMID: 39126207 PMCID: PMC11561673 DOI: 10.1111/acel.14294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
Osteoarthritis (OA) is widely recognized as the prevailing joint disease associated with aging. The ketogenic diet (KD) has been postulated to impede the advancement of various inflammatory ailments. β-Hydroxybutyrate (βOHB), a prominent constituent of ketone bodies, has recently been proposed to possess crucial signaling capabilities. In this study, we propose to explore the role and mechanism of βOHB in OA. Tissue staining and inflammatory factor assay were employed to evaluate the impacts of KD and βOHB on OA rats. The oxidative stress conditions in chondrocytes were induced using tert-butyl hydroperoxide (TBHP). The mechanisms were determined using the siRNA of hydroxycarboxylic acid receptor 2 (HCAR2), the antagonist of adenosine monophosphate-activated protein kinase (AMPK), and the inhibitor of mitophagy. The administration of KD demonstrated a reduction in pathological damage to cartilage, as well as a decrease in plasma levels of inflammatory factors. Furthermore, it resulted in an increase in the concentration of βOHB in the blood and synovial fluid. In vitro experiments showed that βOHB facilitated mitophagy and adenosine triphosphate production. Besides, βOHB mitigated chondrocyte senescence, inflammatory factors secretion, extracellular matrix degradation, and apoptosis induced by TBHP. Subsequent investigations indicated that the protective effects of βOHB were no longer observed following the knockdown of HCAR2, the antagonist of AMPK, or the inhibitor of mitophagy. Moreover, in vivo studies suggested that βOHB played a protective role by targeting the HCAR2-AMPK-PINK1 axis. In conclusion, βOHB enhanced chondrocyte mitophagy through the HCAR2/AMPK/PINK1/Parkin pathway, offering a potential therapeutic approach for the treatment of OA.
Collapse
Affiliation(s)
- Huangming Zhuang
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xunshan Ren
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yuelong Zhang
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Huajie Li
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| | - Panghu Zhou
- Department of OrthopedicsRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
3
|
Farrag M, Cordero-Barreal A, Ait Eldjoudi D, Varela-García M, Torrijos Pulpón C, Lago F, Essawy A, Soffar A, Pino J, Farrag Y, Gualillo O. Impact of Chondrocyte Inflammation on Glial Cell Activation: The Mediating Role of Nitric Oxide. Cartilage 2024:19476035241292323. [PMID: 39469810 PMCID: PMC11556648 DOI: 10.1177/19476035241292323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
OBJECTIVE This study investigates how the inflammatory response of ATDC5 murine chondrogenic cells influences the activity of C6 (rat) and GL261 (mouse) glial cell lines. Prior research suggested nitric oxide (NO) involvement in cartilage-immune crosstalk. The current study explores whether NO, produced by inflamed chondrocytes, mediates signaling between chondrocytes and glial cells. DESIGN Pre-challenged ATDC5 cells with 250 ng/ml of lipopolysaccharide (LPS) were cocultured with GL261 or C6 glioma cells for 24 h with a transwell culture system. Cell viability was assessed using MTT assay. Gene and protein expression were evaluated by qRT-PCR and WB, respectively. RESULTS Real-time reverse transcription-polymerase chain reaction (RT-qPCR) indicated statistically significant upregulation of LCN2, IL-6, TNF-α, IL-1β, and GFAP in glial cells following 24-h coculture with challenged ATDC5 cells. Suppression of LPS-induced NO production by aminoguanidine decreased LPS-mediated LCN2 and IL-6 expression in glioma cells. We identified also the involvement of the ERK1/2 and AKT signaling pathways in the glial neuroinflammatory response. CONCLUSIONS This study demonstrates, for the first time, that NO produced by inflamed murine chondrocytes mediated pro-inflammatory responses in glial cells via ERK1/2 and AKT signaling, highlighting a potential mechanism linking cartilage NO to neuroinflammation and chronic pain in osteoarthritis.
Collapse
Affiliation(s)
- Mariam Farrag
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
- Euro-Mediterranean Master in Neuroscience and Biotechnology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Alfonso Cordero-Barreal
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Djedjiga Ait Eldjoudi
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - María Varela-García
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Carlos Torrijos Pulpón
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Francisca Lago
- Research Laboratory 7, Molecular and Cellular Cardiology Lab, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Amina Essawy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Ahmed Soffar
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Jesus Pino
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Yousof Farrag
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Oreste Gualillo
- Research Laboratory 9, NEIRID Lab Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases, SERGAS Servizo Galego de Saude, IDIS Instituto de Investigación Sanitaria de Santiago, Santiago University Clinical Hospital, Santiago de Compostela, Spain
- Molecular Medicine, International PhD School of University of Santiago EDIUS, University of Santiago, Santiago de Compostela, Spain
| |
Collapse
|
4
|
Farrag Y, Ait Eldjoudi D, Farrag M, González-Rodríguez M, Ruiz-Fernández C, Cordero A, Varela-García M, Torrijos Pulpón C, Bouza R, Lago F, Pino J, Alvarez-Lorenzo C, Gualillo O. Poly(ethylene Glycol) Methyl Ether Methacrylate-Based Injectable Hydrogels: Swelling, Rheological, and In Vitro Biocompatibility Properties with ATDC5 Chondrogenic Lineage. Polymers (Basel) 2023; 15:4635. [PMID: 38139888 PMCID: PMC10747511 DOI: 10.3390/polym15244635] [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: 11/01/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Here, we present the synthesis of a series of chemical homopolymeric and copolymeric injectable hydrogels based on polyethylene glycol methyl ether methacrylate (PEGMEM) alone or with 2-dimethylamino ethyl methacrylate (DMAEM). The objective of this study was to investigate how the modification of hydrogel components influences the swelling, rheological attributes, and in vitro biocompatibility of the hydrogels. The hydrogels' networks were formed via free radical polymerization, as assured by 1H nuclear magnetic resonance spectroscopy (1H NMR). The swelling of the hydrogels directly correlated with the monomer and the catalyst amounts, in addition to the molecular weight of the monomer. Rheological analysis revealed that most of the synthesized hydrogels had viscoelastic and shear-thinning properties. The storage modulus and the viscosity increased by increasing the monomer and the crosslinker fraction but decreased by increasing the catalyst. MTT analysis showed no potential toxicity of the homopolymeric hydrogels, whereas the copolymeric hydrogels were toxic only at high DMEAM concentrations. The crosslinker polyethylene glycol dimethacrylate (PEGDMA) induced inflammation in ATDC5 cells, as detected by the significant increase in nitric oxide synthase type II activity. The results suggest a range of highly tunable homopolymeric and copolymeric hydrogels as candidates for cartilage regeneration.
Collapse
Affiliation(s)
- Yousof Farrag
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Djedjiga Ait Eldjoudi
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Mariam Farrag
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - María González-Rodríguez
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Clara Ruiz-Fernández
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Alfonso Cordero
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - María Varela-García
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Carlos Torrijos Pulpón
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Rebeca Bouza
- Grupo de Polímeros, Departamento de Física y Ciencias de la Tierra, Escuela Universitaria Politécnica, Universidade da Coruña, Serantes, Avda. 19 de Febrero s/n, 15471 Ferrol, Spain;
| | - Francisca Lago
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Molecular and Cellular Cardiology Lab, Research Laboratory 7, Santiago University Clinical Hospital, C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain;
| | - Jesus Pino
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| | - Carmen Alvarez-Lorenzo
- I+D Farma Group (GI-1645), Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Farmacia, Instituto de Materiales (iMATUS), Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Oreste Gualillo
- Servizo Galego de Saude (SERGAS) and Instituto de Investigación Sanitaria de Santiago (IDIS), Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases (NEIRID Group), Santiago University Clinical Hospital, Building C, Travesía da Choupana S/N, 15706 Santiago de Compostela, Spain; (D.A.E.); (M.F.); (M.G.-R.); (C.R.-F.); (A.C.); (M.V.-G.); (C.T.P.); (O.G.)
| |
Collapse
|
5
|
Cruz MA, Gonzalez Y, Vélez Toro JA, Karimzadeh M, Rubbo A, Morris L, Medam R, Splawn T, Archer M, Fernandes RJ, Dennis JE, Kean TJ. Micronutrient optimization for tissue engineered articular cartilage production of type II collagen. Front Bioeng Biotechnol 2023; 11:1179332. [PMID: 37346792 PMCID: PMC10280293 DOI: 10.3389/fbioe.2023.1179332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/23/2023] [Indexed: 06/23/2023] Open
Abstract
Tissue Engineering of cartilage has been hampered by the inability of engineered tissue to express native levels of type II collagen in vitro. Inadequate levels of type II collagen are, in part, due to a failure to recapitulate the physiological environment in culture. In this study, we engineered primary rabbit chondrocytes to express a secreted reporter, Gaussia Luciferase, driven by the type II collagen promoter, and applied a Design of Experiments approach to assess chondrogenic differentiation in micronutrient-supplemented medium. Using a Response Surface Model, 240 combinations of micronutrients absent in standard chondrogenic differentiation medium, were screened and assessed for type II collagen promoter-driven Gaussia luciferase expression. While the target of this study was to establish a combination of all micronutrients, alpha-linolenic acid, copper, cobalt, chromium, manganese, molybdenum, vitamins A, E, D and B7 were all found to have a significant effect on type II collagen promoter activity. Five conditions containing all micronutrients predicted to produce the greatest luciferase expression were selected for further study. Validation of these conditions in 3D aggregates identified an optimal condition for type II collagen promoter activity. Engineered cartilage grown in this condition, showed a 170% increase in type II collagen expression (Day 22 Luminescence) and in Young's tensile modulus compared to engineered cartilage in basal media alone.Collagen cross-linking analysis confirmed formation of type II-type II collagen and type II-type IX collagen cross-linked heteropolymeric fibrils, characteristic of mature native cartilage. Combining a Design of Experiments approach and secreted reporter cells in 3D aggregate culture enabled a high-throughput platform that can be used to identify more optimal physiological culture parameters for chondrogenesis.
Collapse
Affiliation(s)
- Maria A. Cruz
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Yamilet Gonzalez
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Javier A. Vélez Toro
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Makan Karimzadeh
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Anthony Rubbo
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Lauren Morris
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Ramapaada Medam
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Taylor Splawn
- Baylor College of Medicine, Houston, TX, United States
| | - Marilyn Archer
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | - Russell J. Fernandes
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, United States
| | | | - Thomas J. Kean
- Biionix Cluster, Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, United States
- Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
6
|
Piñeiro-Ramil M, Sanjurjo-Rodríguez C, Rodríguez-Fernández S, Hermida-Gómez T, Blanco-García FJ, Fuentes-Boquete I, Vaamonde-García C, Díaz-Prado S. Generation of human immortalized chondrocytes from osteoarthritic and healthy cartilage : a new tool for cartilage pathophysiology studies. Bone Joint Res 2023; 12:46-57. [PMID: 36647698 PMCID: PMC9872042 DOI: 10.1302/2046-3758.121.bjr-2022-0207.r1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
AIMS After a few passages of in vitro culture, primary human articular chondrocytes undergo senescence and loss of their phenotype. Most of the available chondrocyte cell lines have been obtained from cartilage tissues different from diarthrodial joints, and their utility for osteoarthritis (OA) research is reduced. Thus, the goal of this research was the development of immortalized chondrocyte cell lines proceeded from the articular cartilage of patients with and without OA. METHODS Using telomerase reverse transcriptase (hTERT) and SV40 large T antigen (SV40LT), we transduced primary OA articular chondrocytes. Proliferative capacity, degree of senescence, and chondrocyte surface antigen expression in transduced chondrocytes were evaluated. In addition, the capacity of transduced chondrocytes to synthesize a tissue similar to cartilage and to respond to interleukin (IL)-1β was assessed. RESULTS Coexpression of both transgenes (SV40 and hTERT) were observed in the nuclei of transduced chondrocytes. Generated chondrocyte cell lines showed a high proliferation capacity and less than 2% of senescent cells. These cell lines were able to form 3D aggregates analogous to those generated by primary articular chondrocytes, but were unsuccessful in synthesizing cartilage-like tissue when seeded on type I collagen sponges. However, generated chondrocyte cell lines maintained the potential to respond to IL-1β stimulation. CONCLUSION Through SV40LT and hTERT transduction, we successfully immortalized chondrocytes. These immortalized chondrocytes were able to overcome senescence in vitro, but were incapable of synthesizing cartilage-like tissue under the experimental conditions. Nonetheless, these chondrocyte cell lines could be advantageous for OA investigation since, similarly to primary articular chondrocytes, they showed capacity to upregulate inflammatory mediators in response to the IL-1β cytokine.Cite this article: Bone Joint Res 2023;12(1):46-57.
Collapse
Affiliation(s)
- María Piñeiro-Ramil
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain,Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain
| | - Clara Sanjurjo-Rodríguez
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain,Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Silvia Rodríguez-Fernández
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain,Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), A Coruña, Spain
| | - Tamara Hermida-Gómez
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain,Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario da Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain
| | - Francisco J. Blanco-García
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain,Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), A Coruña, Spain,Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario da Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain
| | - Isaac Fuentes-Boquete
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain,Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain,Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), A Coruña, Spain
| | - Carlos Vaamonde-García
- Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Grupo de Investigación en Reumatología (GIR), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario da Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain,Departamento de Biología, Facultad de Ciencias, Universidade da Coruña (UDC), A Coruña, Spain
| | - Silvia Díaz-Prado
- Grupo de Investigación en Terapia Celular y Medicina Regenerativa, Universidade da Coruña (UDC), Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Servizo Galego de Saúde (SERGAS), A Coruña, Spain,Centro de Investigacións Científicas Avanzadas (CICA), Universidade da Coruña (UDC), A Coruña, Spain,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain,Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidade da Coruña (UDC), A Coruña, Spain, Silvia Díaz-Prado. E-mail:
| |
Collapse
|
7
|
Zheng K, Bai J, Yang H, Xu Y, Pan G, Wang H, Geng D. Nanomaterial-assisted theranosis of bone diseases. Bioact Mater 2022; 24:263-312. [PMID: 36632509 PMCID: PMC9813540 DOI: 10.1016/j.bioactmat.2022.12.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 12/27/2022] Open
Abstract
Bone-related diseases refer to a group of skeletal disorders that are characterized by bone and cartilage destruction. Conventional approaches can regulate bone homeostasis to a certain extent. However, these therapies are still associated with some undesirable problems. Fortunately, recent advances in nanomaterials have provided unprecedented opportunities for diagnosis and therapy of bone-related diseases. This review provides a comprehensive and up-to-date overview of current advanced theranostic nanomaterials in bone-related diseases. First, the potential utility of nanomaterials for biological imaging and biomarker detection is illustrated. Second, nanomaterials serve as therapeutic delivery platforms with special functions for bone homeostasis regulation and cellular modulation are highlighted. Finally, perspectives in this field are offered, including current key bottlenecks and future directions, which may be helpful for exploiting nanomaterials with novel properties and unique functions. This review will provide scientific guidance to enhance the development of advanced nanomaterials for the diagnosis and therapy of bone-related diseases.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author.Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Huaiyu Wang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China,Corresponding author.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, Jiangsu, China,Corresponding author. Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
8
|
Trummer M, Galardon E, Mayer B, Steiner G, Stamm T, Kloesch B. Polysulfides derived from the hydrogen sulfide and persulfide donor P* inhibit IL-1β-mediated inducible nitric oxide synthase signaling in ATDC5 cells: are CCAAT/enhancer-binding proteins β and δ involved in the anti-inflammatory effects of hydrogen sulfide and polysulfides? Nitric Oxide 2022; 129:41-52. [DOI: 10.1016/j.niox.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/14/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
|
9
|
Galla R, Ruga S, Ferrari S, Saccone S, Saccuman L, Invernizzi M, Uberti F. In vitro analysis of the effects of plant-derived chondroitin sulfate from intestinal barrier to chondrocytes. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
10
|
Ruiz-Fernández C, González-Rodríguez M, Abella V, Francisco V, Cordero-Barreal A, Ait Eldjoudi D, Farrag Y, Pino J, Conde-Aranda J, González-Gay MÁ, Mera A, Mobasheri A, García-Caballero L, Gándara-Cortés M, Lago F, Scotece M, Gualillo O. WISP-2 modulates the induction of inflammatory mediators and cartilage catabolism in chondrocytes. J Transl Med 2022; 102:989-999. [PMID: 36775427 DOI: 10.1038/s41374-022-00793-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
Wnt-1 inducible signaling pathway protein 2 (WISP-2/CCN5) is a recently identified adipokine that has been described as an important mediator of canonical Wnt activation in adipogenic precursor cells. In osteoarthritis (OA), the most common form of arthritis, chondrocytes exhibit aberrant and increased production of pro-inflammatory mediators and matrix degrading enzymes such as IL-1β and MMP-13. Although recent evidence suggests a role for Wnt signaling in OA physiopathology, little is known about the involvement of WISP-2 in cartilage degradation. In the present study, we determined the expression of WISP-2 in healthy and OA human chondrocytes. WISP-2 expression is modulated along chondrocyte differentiation and downregulated at the onset of hypertrophy by inflammatory mediators. We also investigated the effect of WISP-2 on cartilage catabolism and performed WISP-2 loss-of-function experiments using RNA interference technology in human T/C-28a2 immortalized chondrocytes. We demonstrated that recombinant human WISP-2 protein reduced IL-1β-mediated chondrocyte catabolism, that IL-1β and WNT/b-catenin signaling pathways are involved in rhWISP-2 protein and IL-1β effects in human chondrocytes, and that WISP-2 has a regulatory role in attenuating the catabolic effects of IL-1β in chondrocytes. Gene silencing of WISP-2 increased the induction of the catabolic markers MMP-13 and ADAMTS-5 and the inflammatory mediators IL-6 and IL-8 triggered by IL-1β in human primary OA chondrocytes in a Wnt/β-catenin dependent manner. In conclusion, here we have shown for the first time that WISP-2 may have relevant roles in modulating the turnover of extracellular matrix in the cartilage and that its downregulation may detrimentally alter the inflammatory environment in OA cartilage. We also proved the participation of Wnt/β-catenin signaling pathway in these processes. Thus, targeting WISP-2 might represent a potential therapeutical approach for degenerative and/or inflammatory diseases of musculoskeletal system, such as osteoarthritis.
Collapse
Affiliation(s)
- Clara Ruiz-Fernández
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- International PhD School of the University of Santiago de Compostela (EDIUS), Doctoral Programme in Medicine Clinical Research, Santiago de Compostela, Spain
| | - María González-Rodríguez
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- International PhD School of the University of Santiago de Compostela (EDIUS), Doctoral Programme in Drug Research and Development, Santiago de Compostela, Spain
| | - Vanessa Abella
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Vera Francisco
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Alfonso Cordero-Barreal
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Djedjiga Ait Eldjoudi
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Yousof Farrag
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Jesús Pino
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology Group, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Miguel Ángel González-Gay
- Hospital Universitario Marqués de Valdecilla, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, University of Cantabria, Avenida de Valdecilla s/n, Santander, Cantabria, Spain
| | - Antonio Mera
- SERGAS, Santiago University Clinical Hospital, Division of Rheumatology, Santiago de Compostela, Spain
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics, and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- University Medical Center Utrecht, Departments of Orthopedics, Rheumatology and Clinical Immunology, Utrecht, The Netherlands
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lucía García-Caballero
- Department of Morphological Sciences. School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marina Gándara-Cortés
- Department of Morphological Sciences. School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisca Lago
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Molecular and Cellular Cardiology Lab, Research Laboratory 7, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Morena Scotece
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
11
|
Huang H, Ding X, Xing D, Lin J, Li Z, Lin J. Hyaluronic Acid Oligosaccharide Derivatives Alleviate Lipopolysaccharide-Induced Inflammation in ATDC5 Cells by Multiple Mechanisms. Molecules 2022; 27:5619. [PMID: 36080383 PMCID: PMC9457626 DOI: 10.3390/molecules27175619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/14/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
High molecular weight hyaluronic acids (HMW-HAs) have been used for the palliative treatment of osteoarthritis (OA) for decades, but the pharmacological activity of HA fragments has not been fully explored due to the limited availability of structurally defined HA fragments. In this study, we synthesized a series glycosides of oligosaccharides of HA (o-HAs), hereinafter collectively referred to as o-HA derivatives. Their effects on OA progression were examined in a chondrocyte inflammatory model established by the lipopolysaccharide (LPS)-challenged ATDC5 cells. Cell Counting Kit-8 (CCK-8) assays and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) showed that o-HA derivatives (≤100 μg/mL) exhibited no cytotoxicity and pro-inflammatory effects. We found that the o-HA and o-HA derivatives alleviated LPS-induced inflammation, apoptosis, autophagy and proliferation-inhibition of ATDC5 cells, similar to the activities of HMW-HAs. Moreover, Western blot analysis showed that different HA derivatives selectively reversed the effects of LPS on the expression of extracellular matrix (ECM)-related proteins (MMP13, COL2A1 and Aggrecan) in ATDC5 cells. Our study suggested that o-HA derivatives may alleviate LPS-induced chondrocyte injury by reducing the inflammatory response, maintaining cell proliferation, inhibiting apoptosis and autophagy, and decreasing ECM degradation, supporting a potential oligosaccharides-mediated therapy for OA.
Collapse
Affiliation(s)
- Hesuyuan Huang
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Xuyang Ding
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Jianjing Lin
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| | - Zhongtang Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People’s Hospital, Peking University, Beijing 100044, China
- Arthritis Institute, Peking University, Beijing 100044, China
| |
Collapse
|
12
|
New Hyaluronic Acid from Plant Origin to Improve Joint Protection—An In Vitro Study. Int J Mol Sci 2022; 23:ijms23158114. [PMID: 35897688 PMCID: PMC9332867 DOI: 10.3390/ijms23158114] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Background: In recent decades, hyaluronic acid (HA) has attracted great attention as a new treatment option for osteoarthritis. Classical therapies are not able to stop the cartilage degeneration process nor do they favor tissue repair. Nowadays, it is accepted that high molecular weight HA can reduce inflammation by promoting tissue regeneration; therefore, the aim of this study was to verify the efficacy of a new high molecular weight HA of plant origin (called GreenIuronic®) in maintaining joint homeostasis and preventing the harmful processes of osteoarthritis. Methods: The bioavailability of GreenIuronic® was investigated in a 3D intestinal barrier model that mimics human oral intake while excluding damage to the intestinal barrier. Furthermore, the chemical significance and biological properties of GreenIuronic® were investigated in conditions that simulate osteoarthritis. Results: Our data demonstrated that GreenIuronic® crosses the intestinal barrier without side effects as it has a chemical–biological profile, which could be responsible for many specific chondrocyte functions. Furthermore, in the osteoarthritis model, GreenIuronic® can modulate the molecular mechanism responsible for preventing and restoring the degradation of cartilage. Conclusion: According to our results, this new form of HA appears to be well absorbed and distributed to chondrocytes, preserving their biological activities. Therefore, the oral administration of GreenIuronic® in humans can be considered a valid strategy to obtain beneficial therapeutic effects during osteoarthritis.
Collapse
|
13
|
Qiao Y, Yi D, Reed DA, Mercuri LG, Chen D, Oh CD. A novel approach to establishing a temporomandibular joint fibrocartilage cell line. J Dent Sci 2022; 17:1378-1386. [PMID: 35784155 PMCID: PMC9236962 DOI: 10.1016/j.jds.2022.04.017] [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: 04/18/2022] [Revised: 04/21/2022] [Indexed: 11/21/2022] Open
Abstract
Background/purpose The temporomandibular joint (TMJ) is a bi-arthrodial joint that is composed of the temporal bone glenoid fossa and the condylar head of the mandible both having fibrocartilaginous articular surfaces. Functional overloading of the TMJ is the main cause of TMJ osteoarthritis (TMJ OA) disease. The aim of this study was to establish immortalized TMJ fibrocartilage cell clones to provide enough cells to adequately investigate the molecular mechanisms studies of TMJ OA. Materials and methods We have isolated temporomandibular condyle chondrocytes from adult Sprague Dawley rat. The cells were cultured and immortalized by treating with Y-27632, a well-characterized inhibitor of Rho-Associated Kinase (ROCK). Clones were characterized on the basis of cell morphology and analyses of marker gene expression through 45 passages. Results Cells from the condylar fibrocartilage of the TMJ were successfully immortalized by ROCK inhibitor, retaining a consistent cuboidal cell morphology and the expression of several cell markers of polymorphic cell fate. In addition, they retained phenotype features similar to the primary parental TMJ fibrocartilage cells when the cells were challenged with different cytokines and growth factors. Conclusion These studies establish a novel immortalized cell line through ROCK inhibitor Y-27632, that retains the polymorphic phenotype of primary cell lines from TMJ fibrocartilage chondrocyte cell through a high number of passages, serving as a valuable preclinical resource for mechanistic in vitro assessment of TMJ health, disease, and regeneration.
Collapse
Affiliation(s)
- Yusen Qiao
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, USA
| | - Dan Yi
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, USA
| | - David Andrew Reed
- Department of Oral Biology, University of Illinois Chicago, Chicago, USA
| | - Louis G. Mercuri
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, USA
- Department of Bioengineering, University of Illinois Chicago, Chicago, USA
| | - Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chun-do Oh
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, USA
| |
Collapse
|
14
|
Muenzebrock KA, Kersten V, Alblas J, Garcia JP, Creemers LB. The Added Value of the “Co” in Co-Culture Systems in Research on Osteoarthritis Pathology and Treatment Development. Front Bioeng Biotechnol 2022; 10:843056. [PMID: 35309991 PMCID: PMC8927651 DOI: 10.3389/fbioe.2022.843056] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent disease and a major health burden. Its development and progression are influenced by factors such as age, obesity or joint overuse. As a whole organ disease OA affects not only cartilage, bone and synovium but also ligaments, fatty or nervous tissue surrounding the joint. These joint tissues interact with each other and understanding this interaction is important in developing novel treatments. To incorporate and study these interactions in OA research, several co-culture models have evolved. They combine two or more cell types or tissues and investigate the influence of amongst others inflammatory or degenerative stimuli seen in OA. This review focuses on co-cultures and the differential processes occurring in a given tissue or cell as a consequence of being combined with another joint cell type or tissue, and/or the extent to which a co-culture mimics the in vivo processes. Most co-culture models depart from synovial lining and cartilage culture, but also fat pad and bone have been included. Not all of the models appear to reflect the postulated in vivo OA pathophysiology, although some of the discrepancies may indicate current assumptions on this process are not entirely valid. Systematic analysis of the mutual influence the separate compartments in a given model exert on each other and validation against in vivo or ex vivo observation is still largely lacking and would increase their added value as in vitro OA models.
Collapse
|
15
|
Monomeric C reactive protein (mCRP) regulates inflammatory responses in human and mouse chondrocytes. J Transl Med 2021; 101:1550-1560. [PMID: 33767361 DOI: 10.1038/s41374-021-00584-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
C-reactive protein (CRP) is an acute-phase protein that is used as an established biomarker to follow disease severity and progression in a plethora of inflammatory diseases. However, its pathophysiologic mechanisms of action are still poorly defined and remain elusive. CRP, in its pentameric form, exhibits weak anti-inflammatory activity. On the contrary, the monomeric isoform (mCRP) exhibits potent pro-inflammatory properties in endothelial cells, leukocytes, and platelets. So far, no data exists regarding mCRP effects in human or mouse chondrocytes. This work aimed to verify the pathophysiological relevance of mCRP in the etiology and/or progression of osteoarthritis (OA). We investigated the effects of mCRP in cultured human primary chondrocytes and in the chondrogenic ATDC5 mouse cell line. We determined mRNA and protein levels of relevant factors involved in inflammatory responses and the modulation of nitric oxide synthase type II (NOS2), an early inflammatory molecular target. We demonstrate, for the first time, that monomeric C reactive protein increases NOS2, COX2, MMP13, VCAM1, IL-6, IL-8, and LCN2 expression in human and murine chondrocytes. We also demonstrated that NF-kB is a key factor in the intracellular signaling of mCRP-driven induction of pro-inflammatory and catabolic mediators in chondrocytes. We concluded that mCRP exerts a sustained catabolic effect on human and murine chondrocytes, increasing the expression of inflammatory mediators and proteolytic enzymes, which can promote extracellular matrix (ECM) breakdown in healthy and OA cartilage. In addition, our results implicate the NF-kB signaling pathway in catabolic effects mediated by mCRP.
Collapse
|
16
|
Conde J, Ruiz-Fernandez C, Francisco V, Scotece M, Gómez R, Lago F, Gonzalez-Gay MA, Pino J, Mobasheri A, Gualillo O. Dickkopf-3 (DKK3) Signaling in IL-1α-Challenged Chondrocytes: Involvement of the NF-κB Pathway. Cartilage 2021; 13:925S-934S. [PMID: 32532182 PMCID: PMC8804835 DOI: 10.1177/1947603520933328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is an age-related biomechanical and low-grade inflammometabolic disease of the joints and one of the costliest and disabling forms of arthritis. Studies on matrix-degrading enzymes such as metalloproteases, which are implicated in the increased catabolism of extracellular matrix, are of paramount relevance. DKK3 is a member of DKK family and is best known for its role in cancer. Although there is some information about the participation of DKK3 in cartilage pathophysiology and on metalloproteases regulation, in particular, little is known about DKK3 signaling mechanisms. Thus, the aim of this study is to explore how DKK3 regulates matrix metalloproteinase-13 (MMP-13) expression. DESIGN Gene, protein expression and protein phosphorylation in primary human chondrocytes and ATDC5 mouse cells were assessed by RT-qPCR and Western blot analysis. Further studies on DKK3 activity were performed by targeting DKK3 gene with a specific siRNA. RESULTS DKK3 expression was found to be higher in OA human chondrocytes than healthy cells, being its expression decreased in interleukin-1α (IL-1α)-stimulated cells. DKK3 knockdown increased the induction of MMP-13 elicited by IL-1α in human and mouse chondrocytes and after the analysis of different signalling pathways, we observed that NF-κB pathway was involved in the regulation of MMP-13 expression by DKK3. CONCLUSIONS Herein we have demonstrated, for the first time, that DKK3 gene silencing exacerbated NF-κB activation, resulting in an increased IL-1α-driven induction of MMP-13. Our results further confirm that DKK3 may play a protective role in OA by attenuating NF-κB activation and the subsequent production of metalloproteases.
Collapse
Affiliation(s)
- Javier Conde
- SERGAS (Servizo Galego de Saude) and
IDIS (Instituto de Investigación Sanitaria de Santiago), the NEIRID Lab
(Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago
University Clinical Hospital, Santiago de Compostela, Spain,Javier Conde, The NEIRID Lab, Santiago
University Clinical Hospital, Building C, Level-2, Door 9, Santiago de
Compostela, 15706, Spain.
| | - Clara Ruiz-Fernandez
- SERGAS (Servizo Galego de Saude) and
IDIS (Instituto de Investigación Sanitaria de Santiago), the NEIRID Lab
(Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago
University Clinical Hospital, Santiago de Compostela, Spain
| | - Vera Francisco
- SERGAS (Servizo Galego de Saude) and
IDIS (Instituto de Investigación Sanitaria de Santiago), the NEIRID Lab
(Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago
University Clinical Hospital, Santiago de Compostela, Spain
| | - Morena Scotece
- SERGAS (Servizo Galego de Saude) and
IDIS (Instituto de Investigación Sanitaria de Santiago), the NEIRID Lab
(Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago
University Clinical Hospital, Santiago de Compostela, Spain
| | - Rodolfo Gómez
- Musculoskeletal Pathology Laboratory,
Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela,
Spain
| | - Francisca Lago
- Molecular and Cellular Cardiology Group,
SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de
Santiago), Santiago University Clinical Hospital, Santiago de Compostela,
Spain
| | - Miguel Angel Gonzalez-Gay
- Epidemiology, Genetics and
Atherosclerosis Research Group on Systemic Inflammatory Diseases, Universidad de
Cantabria and IDIVAL, Hospital Universitario Marqués de Valdecilla, Santander,
Spain
| | - Jesús Pino
- SERGAS (Servizo Galego de Saude) and
IDIS (Instituto de Investigación Sanitaria de Santiago), the NEIRID Lab
(Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago
University Clinical Hospital, Santiago de Compostela, Spain
| | - Ali Mobasheri
- Department of Regenerative Medicine,
State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and
IDIS (Instituto de Investigación Sanitaria de Santiago), the NEIRID Lab
(Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago
University Clinical Hospital, Santiago de Compostela, Spain
| |
Collapse
|
17
|
Khella CM, Asgarian R, Horvath JM, Rolauffs B, Hart ML. An Evidence-Based Systematic Review of Human Knee Post-Traumatic Osteoarthritis (PTOA): Timeline of Clinical Presentation and Disease Markers, Comparison of Knee Joint PTOA Models and Early Disease Implications. Int J Mol Sci 2021; 22:1996. [PMID: 33671471 PMCID: PMC7922905 DOI: 10.3390/ijms22041996] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Understanding the causality of the post-traumatic osteoarthritis (PTOA) disease process of the knee joint is important for diagnosing early disease and developing new and effective preventions or treatments. The aim of this review was to provide detailed clinical data on inflammatory and other biomarkers obtained from patients after acute knee trauma in order to (i) present a timeline of events that occur in the acute, subacute, and chronic post-traumatic phases and in PTOA, and (ii) to identify key factors present in the synovial fluid, serum/plasma and urine, leading to PTOA of the knee in 23-50% of individuals who had acute knee trauma. In this context, we additionally discuss methods of simulating knee trauma and inflammation in in vivo, ex vivo articular cartilage explant and in vitro chondrocyte models, and answer whether these models are representative of the clinical inflammatory stages following knee trauma. Moreover, we compare the pro-inflammatory cytokine concentrations used in such models and demonstrate that, compared to concentrations in the synovial fluid after knee trauma, they are exceedingly high. We then used the Bradford Hill Framework to present evidence that TNF-α and IL-6 cytokines are causal factors, while IL-1β and IL-17 are credible factors in inducing knee PTOA disease progresssion. Lastly, we discuss beneficial infrastructure for future studies to dissect the role of local vs. systemic inflammation in PTOA progression with an emphasis on early disease.
Collapse
Affiliation(s)
| | | | | | | | - Melanie L. Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (C.M.K.); (R.A.); (J.M.H.); (B.R.)
| |
Collapse
|
18
|
Haseeb A, Lefebvre V. Isolation of Mouse Growth Plate and Articular Chondrocytes for Primary Cultures. Methods Mol Biol 2021; 2245:39-51. [PMID: 33315194 DOI: 10.1007/978-1-0716-1119-7_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Cartilage is a connective tissue presenting in several forms that are all essential components of the vertebrate skeleton. Complementing in vivo models, cultures of its resident cells-chondrocytes-are important experimental models in mechanistic and preclinical studies relevant to skeletal development and adult homeostasis and to such human pathologies as chondrodysplasias and osteoarthritis. Both growth plate and articular chondrocytes produce pancartilaginous extracellular matrix components, but the two cell subtypes also have distinct phenotypic properties that account for different structural features, functions, and fates of their tissues. Based on study goals, primary chondrocyte cultures should therefore be established from either growth plate or articular cartilage. Here, we describe the methods used in our laboratory to isolate and culture growth plate and articular chondrocytes from neonatal and adult mice, respectively. Both methods involve manual and enzymatic procedures to clean cartilage samples from contaminating tissues and to release chondrocytes as single-cell suspensions from their cartilage matrix.
Collapse
Affiliation(s)
- Abdul Haseeb
- Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Véronique Lefebvre
- Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Volova LT, Pugachev EI, Rossinskaya VV, Boltovskaya VV, Dolgushkin DA, Ossina N. Rheumatoid Arthritis: Applicability of Ready-to-Use Human Cartilaginous Cells for Screening of Compounds with TNF-Alpha Inhibitory Activity. Biomolecules 2020; 10:biom10111563. [PMID: 33212930 PMCID: PMC7698400 DOI: 10.3390/biom10111563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/29/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022] Open
Abstract
In the context of modern drug discovery, there is an obvious advantage to designing phenotypic bioassays based on human disease-relevant cells that express disease-relevant markers. The specific aim of the study was to develop a convenient and reliable method for screening compounds with Tumor Necrosis Factor-alpha (TNF-α) inhibitory activity. This assay was developed using cryopreserved ready-to-use cartilage-derived cells isolated from juvenile donors diagnosed with polydactyly. It has been demonstrated that all donor (10 donors) cells were able to respond to TNF-α treatment by increased secretion of pro-inflammatory cytokine IL-6 into subcultural medium. Inhibition of TNF-α using commercially available TNF-α inhibitor etanercept resulted in a dose-dependent decrease in IL-6 production which was measured by Enzyme-Linked Immunosorbent Assay (ELISA). TNF-α dependent IL-6 production was detected in the cells after both their prolonged cultivation in vitro (≥20 passages) and cryopreservation. This phenotypic bioassay based on ready-to-use primary human cells was developed for detection of novel TNF-α inhibitory compounds and profiling of biosimilar drugs.
Collapse
|
20
|
Song W, Liu Y, Dong X, Song C, Bai Y, Hu P, Li L, Wang T. Lactobacillus M5 prevents osteoarthritis induced by a high-fat diet in mice. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
21
|
K + and Ca 2+ Channels Regulate Ca 2+ Signaling in Chondrocytes: An Illustrated Review. Cells 2020; 9:cells9071577. [PMID: 32610485 PMCID: PMC7408816 DOI: 10.3390/cells9071577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
An improved understanding of fundamental physiological principles and progressive pathophysiological processes in human articular joints (e.g., shoulders, knees, elbows) requires detailed investigations of two principal cell types: synovial fibroblasts and chondrocytes. Our studies, done in the past 8–10 years, have used electrophysiological, Ca2+ imaging, single molecule monitoring, immunocytochemical, and molecular methods to investigate regulation of the resting membrane potential (ER) and intracellular Ca2+ levels in human chondrocytes maintained in 2-D culture. Insights from these published papers are as follows: (1) Chondrocyte preparations express a number of different ion channels that can regulate their ER. (2) Understanding the basis for ER requires knowledge of (a) the presence or absence of ligand (ATP/histamine) stimulation and (b) the extraordinary ionic composition and ionic strength of synovial fluid. (3) In our chondrocyte preparations, at least two types of Ca2+-activated K+ channels are expressed and can significantly hyperpolarize ER. (4) Accounting for changes in ER can provide insights into the functional roles of the ligand-dependent Ca2+ influx through store-operated Ca2+ channels. Some of the findings are illustrated in this review. Our summary diagram suggests that, in chondrocytes, the K+ and Ca2+ channels are linked in a positive feedback loop that can augment Ca2+ influx and therefore regulate lubricant and cytokine secretion and gene transcription.
Collapse
|
22
|
Wu Y, Wang Z, Lin Z, Fu X, Zhan J, Yu K. Salvianolic Acid A Has Anti-Osteoarthritis Effect In Vitro and In Vivo. Front Pharmacol 2020; 11:682. [PMID: 32581777 PMCID: PMC7283387 DOI: 10.3389/fphar.2020.00682] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 04/27/2020] [Indexed: 01/03/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease found in middle-aged and elderly people, which seriously affects their quality of life. The anti-inflammatory and anti-apoptosis pharmacological effects of salvianolic acid A (SAA) have been shown in many studies. In this study, we intended to explore the anti-inflammatory and anti-apoptotic effects of SAA in OA. We evaluated the expression of pro-inflammatory mediators and cartilage matrix catabolic enzymes in chondrocytes by ELISA, Griess reaction, immunofluorescence, and Western blot, which includes nitric oxide (NO), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), prostaglandin E2 (PGE2), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), MMPs (MMP-3, MMP-13), and ADAMTS-5. Bax, Bcl-2, and cleaved caspase-3 were also measured by Western blot methods. The results of this experiment in vitro showed that SAA not only inhibited the production of inflammatory mediators induced by IL-1β and the loss of cartilage matrix but also reduced the apoptosis of mouse chondrocytes induced by IL-1β. According to the results of immunofluorescence and Western blot, SAA inhibited the activation of the NF-κB pathway and MAPK pathway. The results of these in vitro experiments revealed for the first time that SAA down-regulated the production of inflammatory mediators and inhibited the apoptosis of mouse chondrocytes and the degradation of extracellular matrix (ECM), which may be attributed to the inhibition of the activation of NF-κB and MAPK signaling pathways. In the in vivo experiments, 45 mice were randomly divided among three groups (the sham group, OA group, and OA + SAA group). The results of animal experiments showed that SAA treatment for eight consecutive weeks inhibited further deterioration of OA. These results demonstrate that SAA plays an active therapeutic role in the development of OA.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhanghong Wang
- Department of Anesthesiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zeng Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Fu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingdi Zhan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kehe Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
23
|
Mohammadinejad R, Ashrafizadeh M, Pardakhty A, Uzieliene I, Denkovskij J, Bernotiene E, Janssen L, Lorite GS, Saarakkala S, Mobasheri A. Nanotechnological Strategies for Osteoarthritis Diagnosis, Monitoring, Clinical Management, and Regenerative Medicine: Recent Advances and Future Opportunities. Curr Rheumatol Rep 2020; 22:12. [PMID: 32248371 PMCID: PMC7128005 DOI: 10.1007/s11926-020-0884-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW In this review article, we discuss the potential for employing nanotechnological strategies for the diagnosis, monitoring, and clinical management of osteoarthritis (OA) and explore how nanotechnology is being integrated rapidly into regenerative medicine for OA and related osteoarticular disorders. RECENT FINDINGS We review recent advances in this rapidly emerging field and discuss future opportunities for innovations in enhanced diagnosis, prognosis, and treatment of OA and other osteoarticular disorders, the smart delivery of drugs and biological agents, and the development of biomimetic regenerative platforms to support cell and gene therapies for arresting OA and promoting cartilage and bone repair. Nanotubes, magnetic nanoparticles, and other nanotechnology-based drug and gene delivery systems may be used for targeting molecular pathways and pathogenic mechanisms involved in OA development. Nanocomposites are also being explored as potential tools for promoting cartilage repair. Nanotechnology platforms may be combined with cell, gene, and biological therapies for the development of a new generation of future OA therapeutics. Graphical Abstract.
Collapse
Affiliation(s)
- Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania
| | - Lauriane Janssen
- Microelectronics Research Unit, Faculty of Information Technology and Electrical Engineering, University of Oulu, PL 4500, 3FI-90014, Oulu, Finland
| | - Gabriela S Lorite
- Microelectronics Research Unit, Faculty of Information Technology and Electrical Engineering, University of Oulu, PL 4500, 3FI-90014, Oulu, Finland
| | - Simo Saarakkala
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ali Mobasheri
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406, Vilnius, Lithuania.
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland.
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Centre for Sport, Exercise and Osteoarthritis Versus Arthritis, Queen's Medical Centre, Nottingham, UK.
- Sheik Salem Bin Mahfouz Scientific Chair for Treatment of Osteoarthritis with Stem Cells, King AbdulAziz University, Jeddah, Saudi Arabia.
- University Medical Center Utrecht, Department of Orthopedics and Department of Rheumatology & Clinical Immunology, 508 GA, Utrecht, The Netherlands.
| |
Collapse
|
24
|
McDermott BT, Peffers MJ, McDonagh B, Tew SR. Translational regulation contributes to the secretory response of chondrocytic cells following exposure to interleukin-1β. J Biol Chem 2019; 294:13027-13039. [PMID: 31300557 PMCID: PMC6721953 DOI: 10.1074/jbc.ra118.006865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 06/12/2019] [Indexed: 01/18/2023] Open
Abstract
Osteoarthritis is a chronic disease characterized by the loss of articular cartilage in synovial joints through a process of extracellular matrix destruction that is strongly associated with inflammatory stimuli. Chondrocytes undergo changes to their protein translational capacity during osteoarthritis, but a study of how disease-relevant signals affect chondrocyte protein translation at the transcriptomic level has not previously been performed. In this study, we describe how the inflammatory cytokine interleukin 1-β (IL-1β) rapidly affects protein translation in the chondrocytic cell line SW1353. Using ribosome profiling we demonstrate that IL-1β induced altered translation of inflammatory-associated transcripts such as NFKB1, TNFAIP2, MMP13, CCL2, and CCL7, as well as a number of ribosome-associated transcripts, through differential translation and the use of multiple open reading frames. Proteomic analysis of the cellular layer and the conditioned media of these cells identified changes in a number of the proteins that were differentially translated. Translationally regulated secreted proteins included a number of chemokines and cytokines, underlining the rapid, translationally mediated inflammatory cascade that is initiated by IL-1β. Although fewer cellular proteins were found to be regulated in both ribosome profiling and proteomic data sets, we did find increased levels of SOD2, indicative of redox changes within SW1353 cells being modulated at the translational level. In conclusion, we have produced combined ribosome profiling and proteomic data sets that provide a valuable resource in understanding the processes that occur during cytokine stimulation of chondrocytic cells.
Collapse
Affiliation(s)
- Benjamin T McDermott
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom.
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| | - Brian McDonagh
- Department of Physiology, School of Medicine, National University of Ireland (NUI), Galway H91 TK33, Ireland
| | - Simon R Tew
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, United Kingdom
| |
Collapse
|
25
|
Cheleschi S, Calamia V, Fernandez-Moreno M, Biava M, Giordani A, Fioravanti A, Anzini M, Blanco F. In vitro comprehensive analysis of VA692 a new chemical entity for the treatment of osteoarthritis. Int Immunopharmacol 2018; 64:86-100. [DOI: 10.1016/j.intimp.2018.08.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/03/2018] [Accepted: 08/19/2018] [Indexed: 12/21/2022]
|
26
|
Lee KE, Kang YS. l-Citrulline restores nitric oxide level and cellular uptake at the brain capillary endothelial cell line (TR-BBB cells) with glutamate cytotoxicity. Microvasc Res 2018; 120:29-35. [DOI: 10.1016/j.mvr.2018.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/03/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
|
27
|
Baek A, Kim Y, Lee JW, Lee SC, Cho SR. Effect of Polydeoxyribonucleotide on Angiogenesis and Wound Healing in an In Vitro Model of Osteoarthritis. Cell Transplant 2018; 27:1623-1633. [PMID: 30311500 PMCID: PMC6299200 DOI: 10.1177/0963689718804130] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is degenerative disease, leading to pain and functional disability. It is reported that polydeoxyribonucleotide (PDRN) is a suitable therapy for OA. However, the therapeutic mechanisms of PDRN in OA are not fully understood. To investigate the effect of PDRN in an in vitro model of OA, interleukin (IL)-1β or phosphate-buffered saline (PBS) was used to treat a human chondrocytic cell line in hypoxic conditions for 24 h (IL-1β group or control group). PDRN was then used to treat IL-1β group cells for 24 h (PDRN group). By Label-Based Human Antibody Array 1000, angiopoietin-2 (ANG-2), platelet-derived growth factor (PDGF), angiostatin, and endostatin, which were related to angiogenesis, were chosen for further validation studies. Quantitative real-time reverse transcription polymerase chain reaction and western blot analysis validated that the levels of PDGF and ANG-2, which were related to pro-angiogenesis, were significantly increased in the PDRN group compared with those in the control group or the IL-1β group. However, the levels of endostatin and angiostatin, which were related in anti-angiogenesis, were significantly decreased in the PDRN group compared with those in the control group or the IL-1β group. In the same manner, vascular endothelial growth factor, which was a mediator of angiogenesis, was significantly increased in the PDRN group compared with those in the control group or the IL-1β group. Furthermore, wound closure was significantly increased in the PDRN group compared with the control group or the IL-1β group by in vitro scratch assay. Moreover, PDRN decreased expression of metalloproteinase 13, as a catabolic factor for OA, but increased expression of aggrecan, which was an anabolic factor for OA. These data suggest that PDRN may promote angiogenesis and wound healing via down-regulation of catabolism and up-regulation of anabolism in an in vitro model of OA.
Collapse
Affiliation(s)
- Ahreum Baek
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Yoon Kim
- Department of Medicine, The Graduate School of Yonsei University, Seoul, Korea
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Woo Lee
- Department of Orthopedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Chul Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Sang Chul Lee, Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul 03722, South Korea.
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
- Yonsei Stem Cell Center, Avison Biomedical Research Center, Yonsei University College of Medicine, Seoul, South Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, South Korea
- Sung-Rae Cho, Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea.
| |
Collapse
|
28
|
Oxidative stress in osteoarthritis and antioxidant effect of polysaccharide from angelica sinensis. Int J Biol Macromol 2018; 115:281-286. [DOI: 10.1016/j.ijbiomac.2018.04.083] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/25/2018] [Accepted: 04/15/2018] [Indexed: 12/14/2022]
|
29
|
Jin P, Wiraja C, Zhao J, Zhang J, Zheng L, Xu C. Nitric Oxide Nanosensors for Predicting the Development of Osteoarthritis in Rat Model. ACS APPLIED MATERIALS & INTERFACES 2017; 9:25128-25137. [PMID: 28691484 DOI: 10.1021/acsami.7b06404] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Osteoarthritis (OA) is a chronic arthritic disease that causes the overproduction of inflammatory factors such as nitric oxide (NO). This study develops a NO nanosensor to predict the OA development. The nanosensor is synthesized by encapsulating the NO sensing molecules (i.e., 4-amino-5-methylamino-2',7'-difluorofluorescein Diaminofluorescein-FM (DAF-FM)) within the biodegradable poly(lactic-co-glycolic acid) nanoparticles. In vitro, the nanosensor allows the monitoring of the NO release in interleukin-1β-stimulated chondrocytes and the alleviated effect of NG-monomethyl-l-arginine (a NO inhibitor) and andrographolide (an anti-inflammatory agent). In the rat OA model, it permits the quantification of NO level in joint fluid. The proposed NO nanosensor may facilitate a noninvasive and real-time evaluation of the OA development.
Collapse
Affiliation(s)
| | - Christian Wiraja
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University , 50 Nanyang Avenue, 639798 Singapore
| | | | | | | | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University , 70 Nanyang Drive, 637457 Singapore
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University , 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
30
|
Conde J, Lazzaro V, Scotece M, Abella V, Villar R, López V, Gonzalez-Gay MÁ, Pino J, Gómez R, Mera A, Gualillo O. Corticoids synergize with IL-1 in the induction of LCN2. Osteoarthritis Cartilage 2017; 25:1172-1178. [PMID: 28185846 DOI: 10.1016/j.joca.2017.01.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/10/2017] [Accepted: 01/29/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Lipocalin-2 (LCN2) is an adipokine that was first identified in neutrophil granules. In the last years it was recognized as a factor that could impair chondrocyte phenotype, cartilage homeostasis as well as growth plate development. Both pro-inflammatory cytokines and glucocorticoids (GCs) modulate LCN2 expression. Actually, GCs were found to be LCN2 inducers, suggesting that part of the negative actions exerted by these anti-inflammatory drugs at cartilage level could be mediated by this adipokine. So, in this study we wanted to investigate whether corticoids were able to act in synergy with IL-1 in the induction of LCN2 and the signaling pathway involved in this process. MATERIALS AND METHODS For the realization of this work, ATDC5 mouse chondrogenic cell line was used. We determined the mRNA and protein expression of LCN2 by real-time reverse transcription-polymerase chain reaction (RT-qPCR) and western blot respectively, after GC or mineralcorticoid treatment. Different signaling pathways inhibitors were also used. RESULTS GC and mineralcorticoid were able to induce the expression of LCN2 in ATDC5 cells. Interestingly, both corticoids synergized with IL-1 in the induction of LCN2. The effect of these corticoids on the expression of LCN2 occurred through GC or mineralcorticoid receptors and the kinases PI3K, ERK1/2 and JAK2. CONCLUSIONS Prolonged use of corticoids may have detrimental effects on cartilage homeostasis. Based on our results, we conclude that corticoids could increase the negative actions exerted by IL-1 by increasing the expression of LCN2.
Collapse
Affiliation(s)
- J Conde
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| | - V Lazzaro
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - M Scotece
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - V Abella
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - R Villar
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - V López
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - M Á Gonzalez-Gay
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, Universidad de Cantabria and IDIVAL, Santander, Spain
| | - J Pino
- SERGAS (Servizo Galego de Saude), Division of Orthopaedics Surgery and Traumatology, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - R Gómez
- Musculoskeletal Pathology Laboratory, Institute IDIS, Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - A Mera
- SERGAS (Servizo Galego de Saude), Division of Rheumatology, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - O Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
31
|
Phosphate regulates chondrogenesis in a biphasic and maturation-dependent manner. Differentiation 2017; 95:54-62. [PMID: 28511052 DOI: 10.1016/j.diff.2017.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 04/18/2017] [Accepted: 04/27/2017] [Indexed: 01/05/2023]
Abstract
Inorganic phosphate (Pi) has been recognized as an important signaling molecule that modulates chondrocyte maturation and cartilage mineralization. However, conclusive experimental evidence for its involvement in early chondrogenesis is still lacking. Here, using high-density monolayer (2D) and pellet (3D) culture models of chondrogenic ATDC5 cells, we demonstrate that the cell response to Pi does not correlate with the Pi concentration in the culture medium but is better predicted by the availability of Pi on a per cell basis (Pi abundance). Both culture models were treated with ITS+, 10mM β-glycerophosphate (βGP), or ITS+/10mM βGP, which resulted in three levels of Pi abundance in cultures: basal (Pi/DNA <10ng/µg), moderate (Pi/DNA=25.3 - 32.3ng/µg), and high abundance (Pi/DNA >60ng/µg). In chondrogenic medium alone, the abundance levels were at the basal level in 2D culture and moderate in 3D cultures. The addition of 10mM βGP resulted in moderate abundance in 2D and high abundance in 3D cultures. Moderate Pi abundance enhanced early chondrogenesis and production of aggrecan and type II collagen whereas high Pi abundance inhibited chondrogenic differentiation and induced rapid mineralization. Inhibition of sodium phosphate transporters reduced phosphate-induced expression of chondrogenic markers. When 3D ITS+/βGP cultures were treated with levamisole to reduce ALP activity, Pi abundance was decreased to moderate levels, which resulted in significant upregulation of chondrogenic markers, similar to the response in 2D cultures. Delay of phosphate delivery until after early chondrogenesis occurs (7 days) no longer enhanced chondrogenesis, but instead accelerated hypertrophy and mineralization. Together, our data highlights the dependence of chondroprogenitor cell response to Pi on its availability to individual cells and the chondrogenic maturation stage of these cells and suggest that appropriate temporal delivery of phosphate to ATDC5 cells in 3D cultures represents a rapid model for mechanistic studies into the effects of exogenous cues on chondrogenic differentiation, chondrocyte maturation, and matrix mineralization.
Collapse
|
32
|
Tun T, Kang YS. Effects of simvastatin on CAT-1-mediated arginine transport and NO level under high glucose conditions in conditionally immortalized rat inner blood-retinal barrier cell lines (TR-iBRB). Microvasc Res 2017; 111:60-66. [DOI: 10.1016/j.mvr.2017.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 01/05/2017] [Accepted: 01/07/2017] [Indexed: 02/07/2023]
|
33
|
Gao LN, Feng QS, Zhang XF, Wang QS, Cui YL. Tetrandrine suppresses articular inflammatory response by inhibiting pro-inflammatory factors via NF-κB inactivation. J Orthop Res 2016; 34:1557-68. [PMID: 26748661 DOI: 10.1002/jor.23155] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/29/2015] [Indexed: 02/04/2023]
Abstract
Targeting activated macrophages using anti-inflammatory phytopharmaceuticals has been proposed as general therapeutic approaches for rheumatic diseases. Besides macrophages, chondrocytes are another promising target of anti-inflammatory agents. Tetrandrine is a major bisbenzylisoquinoline alkaloid isolated from Stephania tetrandrae S. Moore which has been used for 2,000 years as an antirheumatic herbal drug in China. Although, the anti-inflammatory effect of tetrandrine has been demonstrated, the mechanism has not been clearly clarified. In this study, we designed a comprehensive anti-inflammatory evaluation system for tetrandrine, including complete Freund's adjuvant (CFA)-induced arthritis rat, LPS-induced macrophage RAW 264.7 cells, and chondrogenic ATDC5 cells. The results showed that tetrandrine alleviated CFA-induced foot swelling, synovial inflammation, and pro-inflammatory cytokines secretion. Tetrandrine could inhibit IL-6, IL-1β, and TNF-α expression via blocking the nuclear translocation of nuclear factor (NF)-κB p65 in LPS-induced RAW 264.7 cells. Moreover, ATDC5 cells well responded to LPS induced pro-inflammatory mediators secretion and tissue degradation, and tetrandrine could also inhibit the production of nitric oxide and prostaglandin E2 , as well as the expression of matrix metalloproteinase (MMP)-3 and tissue inhibitor of metalloproteinase (TIMP)-1 via inhibiting IκBα phosphorylation and degradation. In conclusion, the results showed that one of the anti-inflammatory mechanisms of tetrandrine was inhibiting IκBα and NF-κB p65 phosphorylation in LPS-induced macrophage RAW 264.7 cells and chondrogenic ATDC5 cells. Moreover, we introduce a vigorous in vitro cell screening system, LPS-induced murine macrophage RAW 264.7 cells coupling chondrogenic ADTC5 cells, for screening anti-rheumatic drugs. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1557-1568, 2016.
Collapse
Affiliation(s)
- Li-Na Gao
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China
| | - Qi-Shuai Feng
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,The Institute for Biomedical Engineering and Nano Science, Tongji University School of Medicine, 67 Chifeng Road, Shanghai, 200092, China
| | - Xin-Fang Zhang
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China
| | - Qiang-Song Wang
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, No. 236 BaiDi Road, Nankai District, Tianjin, 300192, China
| | - Yuan-Lu Cui
- Research Center of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, No. 88 YuQuan Road, Nankai District, Tianjin, 300193, China
| |
Collapse
|
34
|
Conde J, Otero M, Scotece M, Abella V, López V, Pino J, Gómez R, Lago F, Goldring MB, Gualillo O. E74-like factor 3 and nuclear factor-κB regulate lipocalin-2 expression in chondrocytes. J Physiol 2016; 594:6133-6146. [PMID: 27222093 DOI: 10.1113/jp272240] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022] Open
Abstract
KEY POINTS E74-like factor 3 (ELF3) is a transcription factor regulated by inflammation in different physio-pathological situations. Lipocalin-2 (LCN2) emerged as a relevant adipokine involved in the regulation of inflammation. In this study we showed for the first time the involvement of ELF3 in the control of LCN2 expression and its cooperation with nuclear factor-κB (NFκB). Our results will help to better understand of the role of ELF3, NFκB and LCN2 in the pathophysiology of articular cartilage. ABSTRACT E74-like factor 3 (ELF3) is a transcription factor induced by inflammatory cytokines in chondrocytes that increases gene expression of catabolic and inflammatory mediators. Lipocalin 2 (LCN2) is a novel adipokine that negatively impacts articular cartilage, triggering catabolic and inflammatory responses in chondrocytes. Here, we investigated the control of LCN2 gene expression by ELF3 in the context of interleukin 1 (IL-1)-driven inflammatory responses in chondrocytes. The interaction of ELF3 and nuclear factor-κB (NFκB) in modulating LCN2 levels was also explored. LCN2 mRNA and protein levels, as well those of several other ELF3 target genes, were determined by RT-qPCR and Western blotting. Human primary chondrocytes, primary chondrocytes from wild-type and Elf3 knockout mice, and immortalized human T/C-28a2 and murine ATDC5 cell lines were used in in vitro assays. The activities of various gene reporter constructs were evaluated by luciferase assays. Gene overexpression and knockdown were performed using specific expression vectors and siRNA technology, respectively. ELF3 overexpression transactivated the LCN2 promoter and increased the IL-1-induced mRNA and protein levels of LCN2, as well as the mRNA expression of other pro-inflammatory mediators, in human and mouse chondrocytes. We also identified a collaborative loop between ELF3 and NFκB that amplifies the induction of LCN2. Our findings show a novel role for ELF3 and NFκB in the induction of the pro-inflammatory adipokine LCN2, providing additional evidence of the interaction between ELF3 and NFκB in modulating inflammatory responses, and a better understanding of the mechanisms of action of ELF3 in chondrocytes.
Collapse
Affiliation(s)
- Javier Conde
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Miguel Otero
- Tissue Engineering Regeneration and Repair Program, The Hospital for Special Surgery, and Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Morena Scotece
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Vanessa Abella
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Verónica López
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Jesús Pino
- SERGAS (Servizo Gallego de Saude), Santiago University Clinical Hospital, Division of Orthopaedic Surgery, Santiago de Compostela, Spain
| | - Rodolfo Gómez
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Francisca Lago
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain
| | - Mary B Goldring
- Tissue Engineering Regeneration and Repair Program, The Hospital for Special Surgery, and Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, The NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Santiago de Compostela, 15706, Spain.
| |
Collapse
|
35
|
Carmona JU, Ríos DL, López C, Álvarez ME, Pérez JE, Bohórquez ME. In vitro effects of platelet-rich gel supernatants on histology and chondrocyte apoptosis scores, hyaluronan release and gene expression of equine cartilage explants challenged with lipopolysaccharide. BMC Vet Res 2016; 12:135. [PMID: 27369779 PMCID: PMC4929746 DOI: 10.1186/s12917-016-0759-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 06/23/2016] [Indexed: 01/05/2023] Open
Abstract
Background Platelet-rich plasma (PRP) preparations are a common treatment in equine osteoarthritis (OA). However, there are controversies regarding the ideal concentration of platelets and leukocytes in these biological substances necessary to induce an adequate anti-inflammatory and anabolic response in articular cartilage. The aims were to study the influence of leukocyte- and platelet-rich gel (L-PRG) and pure platelet-rich gel (P-PRG) supernatants on the histological changes of cartilage, the degree of chondrocyte apoptosis, the production of hyaluronan (HA) and the gene expression of nuclear factor kappa beta (NFkβ), matrix metalloproteinase 13 (MMP-13), a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS-4), collagen type I alpha 1 (COL1A1), collagen type II alpha 1 (COL2A1) and cartilage oligomeric matrix protein (COMP) in normal cartilage explants (CEs) challenged with lipopolysaccharide (LPS). Results Overall, 25 % L-PRG supernatant (followed in order of importance by, 50 % P-PRG, 25 % P-PRG and 50 % L-PRG) represented the substance with the most important anti-inflammatory and anabolic effect. 25 % P-PRG supernatant presented important anabolic effects, but it induced a more severe chondrocyte apoptosis than the other evaluated substances. Conclusions 25 % L-PRG supernatant presented the best therapeutic profile. Our results demonstrate that the biological variability of PRP preparations makes their application rather challenging. Additional in vivo research is necessary to know the effect of PRP preparations at different concentrations.
Collapse
Affiliation(s)
- Jorge U Carmona
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Manizales, Colombia.
| | - Diana L Ríos
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Manizales, Colombia
| | - Catalina López
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Manizales, Colombia
| | - María E Álvarez
- Grupo de Investigación Terapia Regenerativa, Departamento de Salud Animal, Universidad de Caldas, Manizales, Colombia.,Grupo de investigación Biosalud, Departamento de Ciencias Básicas para la Salud, Universidad de Caldas, Manizales, Colombia
| | - Jorge E Pérez
- Grupo de investigación Biosalud, Departamento de Ciencias Básicas para la Salud, Universidad de Caldas, Manizales, Colombia
| | - Mabel E Bohórquez
- Grupo de Investigación en Citogenética, Filogenia y Evolución de Poblaciones, Universidad del Tolima, Ibagué, Colombia
| |
Collapse
|
36
|
Patent highlights October–November 2015. Pharm Pat Anal 2016. [DOI: 10.4155/ppa.15.47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical R&D.
Collapse
|
37
|
The novel adipokine progranulin counteracts IL-1 and TLR4-driven inflammatory response in human and murine chondrocytes via TNFR1. Sci Rep 2016; 6:20356. [PMID: 26853108 PMCID: PMC4745010 DOI: 10.1038/srep20356] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022] Open
Abstract
Progranulin (PGRN) is a recently identified adipokine that is supposed to have anti-inflammatory actions. The proinflammatory cytokine interleukin-1β (IL1β) stimulates several mediators of cartilage degradation. Toll like receptor-4 (TLR4) can bind to various damage-associated molecular patterns, leading to inflammatory condition. So far, no data exist of PGRN effects in inflammatory conditions induced by IL1β or lipopolysaccharide (LPS). Here, we investigated the anti-inflammatory potential of PGRN in IL1β- or LPS-induced inflammatory responses of chondrocytes. Human osteoarthritic chondrocytes and ATDC-5 cells were treated with PGRN in presence or not of IL1β or LPS. First, we showed that recombinant PGRN had no effects on cell viability. We present evidence that PGRN expression was increased during the differentiation of ATDC-5 cell line. Moreover, PGRN mRNA and protein expression is increased in cartilage, synovial and infrapatellar fat pad tissue samples from OA patients. PGRN mRNA levels are upregulated under TNFα and IL1β stimulation. Our data showed that PGRN is able to significantly counteract the IL1β-induced expression of NOS2, COX2, MMP13 and VCAM-1. LPS-induced expression of NOS2 is also decreased by PGRN. These effects are mediated, at least in part, through TNFR1. Taken together, our results suggest that PGRN has a clear anti-inflammatory function.
Collapse
|