1
|
Ma W, Tan X, Xie Z, Yu J, Li P, Lin X, Ouyang S, Liu Z, Hou Q, Xie N, Peng T, Li L, Dai Z, Chen X, Xie W. P53: A Key Target in the Development of Osteoarthritis. Mol Biotechnol 2024; 66:1-10. [PMID: 37154864 DOI: 10.1007/s12033-023-00736-9] [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: 11/29/2022] [Accepted: 03/25/2023] [Indexed: 05/10/2023]
Abstract
Osteoarthritis (OA), a chronic degenerative disease characterized mainly by damage to the articular cartilage, is increasingly relevant to the pathological processes of senescence, apoptosis, autophagy, proliferation, and differentiation of chondrocytes. Clinical strategies for osteoarthritis can only improve symptoms and even along with side effects due to age, sex, disease, and other factors. Therefore, there is an urgent need to identify new ideas and targets for current clinical treatment. The tumor suppressor gene p53, which has been identified as a potential target for tumor therapeutic intervention, is responsible for the direct induction of the pathological processes involved in OA modulation. Consequently, deciphering the characteristics of p53 in chondrocytes is essential for investigating OA pathogenesis due to p53 regulation in an array of signaling pathways. This review highlights the effects of p53 on senescence, apoptosis, and autophagy of chondrocytes and its role in the development of OA. It also elucidates the underlying mechanism of p53 regulation in OA, which may help provide a novel strategies for the clinical treatment of OA.
Collapse
Affiliation(s)
- Wentao Ma
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoqian Tan
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhongcheng Xie
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Jiang Yu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Pin Li
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Xiaoyan Lin
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Ouyang
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhiyang Liu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Qin Hou
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Nan Xie
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Tianhong Peng
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Liang Li
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Zhu Dai
- Department of Orthopedics, Hengyang Medical School, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China.
| | - Xi Chen
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
Lin J, Wu G, Chen J, Fu C, Hong X, Li L, Liu X, Wu M. Electroacupuncture inhibits sodium nitroprusside‑mediated chondrocyte apoptosis through the mitochondrial pathway. Mol Med Rep 2018; 18:4922-4930. [PMID: 30272266 PMCID: PMC6236293 DOI: 10.3892/mmr.2018.9498] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022] Open
Abstract
In China, electroacupuncture (EA) is a therapeutic method that is extensively applied in the clinical treatment of osteoarthritis (OA); however, the underlying molecular mechanism remains unclear. Chondrocyte apoptosis may be observed in cartilage tissue in OA, and is often considered a key target for the treatment of this condition. Therefore, the present study aimed to determine the effects of EA on sodium nitroprusside (SNP)-induced chondrocyte apoptosis. Chondrocytes were obtained from the knee joints of Sprague Dawley rats by type II collagenase digestion. Following microscopic observation and authentication with type II collagen immunohistochemistry, articular cartilage cells were used in subsequent experiments. Using inverted phase contrast microscopy, DAPI staining and flow cytometry, it was revealed that chondrocytes treated with SNP became apoptotic, whereas EA inhibited SNP-induced chondrocyte apoptosis. Subsequently, JC-1 single staining, reverse transcription-quantitative polymerase chain reaction analysis, western blotting, colorimetric assays and immunofluorescence staining were performed for further investigation. The results demonstrated that, when compared with normal chondrocytes, the mitochondrial membrane potential of SNP-treated chondrocytes was markedly lowered, B-cell lymphoma 2 (Bcl-2) expression was reduced, and the expression levels of Bcl-2-associated X protein (Bax), cytochrome c, caspase-9 and caspase-3 were increased. Compared with in SNP-treated chondrocytes, the decrease in the mitochondrial membrane potential of chondrocytes treated with SNP and EA was smaller, Bcl-2 expression was increased, and the expression levels of Bax, cytochrome c, caspase-9 and caspase-3 were decreased following EA intervention. In conclusion, the present study demonstrated that EA modulated the mitochondrial pathway to suppress SNP-mediated chondrocyte apoptosis. Therefore, EA may be of value in the treatment of OA.
Collapse
Affiliation(s)
- Jie Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Guangwen Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Changlong Fu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiue Hong
- Department of Acupuncture and Moxibusion, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Li Li
- Department of Acupuncture and Moxibusion, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Xianxiang Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Mingxia Wu
- Department of Acupuncture and Moxibusion, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| |
Collapse
|
3
|
Lee M, Rey K, Besler K, Wang C, Choy J. Immunobiology of Nitric Oxide and Regulation of Inducible Nitric Oxide Synthase. Results Probl Cell Differ 2017; 62:181-207. [PMID: 28455710 DOI: 10.1007/978-3-319-54090-0_8] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is a bioactive gas that has multiple roles in innate and adaptive immune responses. In macrophages, nitric oxide is produced by inducible nitric oxide synthase upon microbial and cytokine stimulation. It is needed for host defense against pathogens and for immune regulation. This review will summarize the role of NO and iNOS in inflammatory and immune responses and will discuss the regulatory mechanisms that control inducible nitric oxide synthase expression and activity.
Collapse
Affiliation(s)
- Martin Lee
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Kevin Rey
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Katrina Besler
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Christine Wang
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Jonathan Choy
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
4
|
Vaamonde-Garcia C, Courties A, Pigenet A, Laiguillon MC, Sautet A, Houard X, Kerdine-Römer S, Meijide R, Berenbaum F, Sellam J. The nuclear factor-erythroid 2-related factor/heme oxygenase-1 axis is critical for the inflammatory features of type 2 diabetes-associated osteoarthritis. J Biol Chem 2017; 292:14505-14515. [PMID: 28684418 DOI: 10.1074/jbc.m117.802157] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/05/2017] [Indexed: 12/11/2022] Open
Abstract
Epidemiological findings support the hypothesis that type 2 diabetes mellitus (T2DM) is a risk factor for osteoarthritis (OA). Moreover, OA cartilage from patients with T2DM exhibits a greater response to inflammatory stress, but the molecular mechanism is unclear. To investigate whether the antioxidant defense system participates in this response, we examined here the expression of nuclear factor-erythroid 2-related factor (Nrf-2), a master antioxidant transcription factor, and of heme oxygenase-1 (HO-1), one of its main target genes, in OA cartilage from T2DM and non-T2DM patients as well as in murine chondrocytes exposed to high glucose (HG). Ex vivo experiments indicated that Nrf-2 and HO-1 expression is reduced in T2DM versus non-T2DM OA cartilage (0.57-fold Nrf-2 and 0.34-fold HO-1), and prostaglandin E2 (PGE2) release was increased in samples with low HO-1 expression. HG-exposed, IL-1β-stimulated chondrocytes had lower Nrf-2 levels in vitro, particularly in the nuclear fraction, than chondrocytes exposed to normal glucose (NG). Accordingly, HO-1 levels were also decreased (0.49-fold) in these cells. The HO-1 inducer cobalt protoporphyrin IX more efficiently attenuated PGE2 and IL-6 release in HG+IL-1β-treated cells than in NG+IL-1β-treated cells. Greater reductions in HO-1 expression and increase in PGE2/IL-6 production were observed in HG+IL-1β-stimulated chondrocytes from Nrf-2-/- mice than in chondrocytes from wild-type mice. We conclude that the Nrf-2/HO-1 axis is a critical pathway in the hyperglucidic-mediated dysregulation of chondrocytes. Impairments in this antioxidant system may explain the greater inflammatory responsiveness of OA cartilage from T2DM patients and may inform treatments of such patients.
Collapse
Affiliation(s)
- Carlos Vaamonde-Garcia
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France.,Tissue Engineering and Cellular Therapy Group, Department of Medicine, University of A Coruña, 15006 A Coruña, Spain
| | - Alice Courties
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France.,Department of Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, 75012 Paris, France
| | - Audrey Pigenet
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France
| | - Marie-Charlotte Laiguillon
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France
| | - Alain Sautet
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Department of Orthopedic Surgery, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, 75012 Paris, France, and
| | - Xavier Houard
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France
| | - Saadia Kerdine-Römer
- INSERM UMR 996, University of Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Rosa Meijide
- Tissue Engineering and Cellular Therapy Group, Department of Medicine, University of A Coruña, 15006 A Coruña, Spain
| | - Francis Berenbaum
- From the Sorbonne University, UPMC University of Paris 06, Paris, France, .,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France.,Department of Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, 75012 Paris, France
| | - Jérémie Sellam
- From the Sorbonne University, UPMC University of Paris 06, Paris, France.,Saint Antoine Medical Faculty, INSERM UMR_S938, 75012 Paris, France.,Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Saint-Antoine Hospital, 75012 Paris, France.,Department of Rheumatology, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, 75012 Paris, France
| |
Collapse
|
5
|
Preconditioning is hormesis part I: Documentation, dose-response features and mechanistic foundations. Pharmacol Res 2016; 110:242-264. [DOI: 10.1016/j.phrs.2015.12.021] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/16/2022]
|
6
|
Quan YY, Qin GQ, Huang H, Liu YH, Wang XP, Chen TS. Dominant roles of Fenton reaction in sodium nitroprusside-induced chondrocyte apoptosis. Free Radic Biol Med 2016; 94:135-44. [PMID: 26923801 DOI: 10.1016/j.freeradbiomed.2016.02.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 12/14/2015] [Accepted: 02/22/2016] [Indexed: 01/08/2023]
Abstract
Sodium nitroprusside (SNP) has been widely used as an exogenous nitric oxide (NO) donor to explore the molecular mechanism of NO-mediated chondrocyte apoptosis during the latest two decades. We have recently found that NO-independent ROS play a key role in SNP-induced apoptosis in rabbit chondrocytes. This study aims to investigate what kind of ROS and how the reliable ROS mediators mediate the SNP-induced apoptosis. Data shows that SNP and NO-exhausted SNP (SNPex) induced ROS production or cytotoxicity to identically degree. SNP induced a marked increase in iron ions, superoxide anion (O2(•-)), hydrogen peroxide (H2O2) and hydroxyl radical ((•)OH) level. H2O2 scavenger (CAT) and (•)OH scavenger (DMSO) significantly inhibited SNP-induced chondrocyte apoptosis. Iron ions chelator (DFO) entirely prevented SNP-induced chondrocyte apoptosis. In contrast, O2(•-) scavenger (SOD) and glutathione depletion agent (BSO) promoted SNP-induced cytotoxicity. K3[Fe(CN)6] exhibited no cytotoxicity, and H2O2 alone up to 250µM or iron ions alone up to 90µM is non-cytotoxic to chondrocytes. Combination of 25µM FeSO4 and 100µM H2O2 in the presence of BSO induced chondrocyte death similar to SNP treatment. Fetal bovine serum (FBS) enhanced iron ions release from SNP and the cytotoxicity of SNP. Our data shows that the extracellular Fenton reaction between iron ions released from SNP and H2O2 induced by SNP plays a key role in SNP-induced chondrocyte apoptosis. Overall, our results indicate that the potential of SNP to increase iron ions and ROS should be especially considered for some biological functions and, possibly, also for clinical applications of this drug.
Collapse
Affiliation(s)
- Ying-Yao Quan
- Department of Pain Management, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Gui-Qi Qin
- MOE Key Laboratory of Laser Life Science & College of Life Science, South China Normal University, Guangzhou, China
| | - Hao Huang
- Department of Pain Management, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yu-Hong Liu
- Department of Pain Management, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Ping Wang
- Department of Pain Management, the First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Tong-Sheng Chen
- MOE Key Laboratory of Laser Life Science & College of Life Science, South China Normal University, Guangzhou, China.
| |
Collapse
|
7
|
Chen Q, Mei X, Han G, Ling P, Guo B, Guo Y, Shao H, Wang G, Cui Z, Bai Y, Xu F. Xanthan gum protects rabbit articular chondrocytes against sodium nitroprusside-induced apoptosis in vitro. Carbohydr Polym 2015; 131:363-9. [PMID: 26256195 DOI: 10.1016/j.carbpol.2015.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/22/2015] [Accepted: 06/01/2015] [Indexed: 01/01/2023]
Abstract
We have previously reported that intra-articular injection of xanthan gum (XG) could significantly ameliorate the degree of joint cartilage degradation and pain in experimental osteoarthritis (OA) model in vivo. In this present study, we evaluated the protective effect of XG against Sodium nitroprusside (SNP)-induced rabbit articular chondrocytes apoptosis in vitro. Rabbit articular chondrocytes were incubated with various concentrations of XG for 24h prior to 0.5mmol/L SNP co-treatment for 24h. The proliferation of chondrocytes was analyzed using MTT assay. The chondrocytes early apoptosis rates were evaluated using Annexin V-FITC/PI flow cytometry. The morphology of apoptosis chondrocytes were observed by scanning electron microscopy (SEM). The loss/disruption of mitochondrial membrane potential was detected using rhodamin 123 by confocal microscope. The concentration of prostaglandin E2 (PGE2) in cell culture supernatants was evaluated using ELISA assay. The results showed that XG could significantly reverse SNP-reduced cell proliferation and inhibited cell early apoptosis rate in a dose-dependent manner. XG alleviated loss/disruption of mitochondrial membrane potential and decreased the PGE2 level of chondrocytes cell culture supernatants in SNP-induced chondrocytes. These results of the present research strongly suggest that XG can protect rabbit articular chondrocytes against SNP-induced apoptosis in vitro.
Collapse
Affiliation(s)
- Qixin Chen
- School of Pharmaceutical Sciences, Liaoning Medical University, Jinzhou 121001, China
| | - Xifan Mei
- The First Affiliated Hospital of Liaoning Medical University, Jinzhou 121001, China
| | - Guanying Han
- The First Affiliated Hospital of Liaoning Medical University, Jinzhou 121001, China; Post-doctoral Scientific Research Workstation, Institute of Biopharmaceuticals of Shandong Province, Jinan 250101, China; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Peixue Ling
- Post-doctoral Scientific Research Workstation, Institute of Biopharmaceuticals of Shandong Province, Jinan 250101, China
| | - Bin Guo
- The First Affiliated Hospital of Liaoning Medical University, Jinzhou 121001, China
| | - Yuewei Guo
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huarong Shao
- Post-doctoral Scientific Research Workstation, Institute of Biopharmaceuticals of Shandong Province, Jinan 250101, China
| | - Guan Wang
- School of Pharmaceutical Sciences, Liaoning Medical University, Jinzhou 121001, China
| | - Zan Cui
- The First Affiliated Hospital of Liaoning Medical University, Jinzhou 121001, China
| | - Yuxin Bai
- School of Pharmaceutical Sciences, Liaoning Medical University, Jinzhou 121001, China
| | - Fang Xu
- School of Pharmaceutical Sciences, Liaoning Medical University, Jinzhou 121001, China
| |
Collapse
|
8
|
The characteristics of thrombin in osteoarthritic pathogenesis and treatment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:407518. [PMID: 25313362 PMCID: PMC4182002 DOI: 10.1155/2014/407518] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/01/2014] [Indexed: 01/08/2023]
Abstract
Osteoarthritis (OA) is a mechanical abnormality associated with degradation of joints. It is characterized by chronic, progressive degeneration of articular cartilage, abnormalities of bone, and synovial change. The most common symptom of OA is local inflammation resulting from exogenous stress or endogenous abnormal cytokines. Additionally, OA is associated with local and/or systemic activation of coagulation and anticoagulation pathways. Thrombin plays an important role in the stimulation of fibrin deposition and the proinflammatory processes in OA. Thrombin mediates hemostatic and inflammatory responses and guides the immune response to tissue damage. Thrombin activates intracellular signaling pathways by interacting with transmembrane domain G protein coupled receptors (GPCRs), known as protease-activated receptors (PARs). In pathogenic mechanisms, PARs have been implicated in the development of acute and chronic inflammatory responses in OA. Therefore, discovery of thrombin signaling pathways would help us to understand the mechanism of OA pathogenesis and lead us to develop therapeutic drugs in the future.
Collapse
|
9
|
Kim KM, Park SE, Lee MS, Kim K, Park YC. Induction of heme oxygenase‑1 expression protects articular chondrocytes against cilostazol‑induced cellular senescence. Int J Mol Med 2014; 34:1335-40. [PMID: 25175370 DOI: 10.3892/ijmm.2014.1918] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/25/2014] [Indexed: 11/06/2022] Open
Abstract
Chondrocyte senescence is associated with the aging and degeneration of cartilage, and eventually leads to joint destruction. The aim of this study was to elucidate the mechanisms responsible for the cytoprotective effects of heme oxygenase‑1 (HO‑1) on chondrocytes in cartilage. Chondrocyte senescence was induced using cilostazol and measured using a specific senescence‑associated β‑galactosidase (SA‑β‑gal) staining assay. Cilostazol altered the expression of type Ⅱ collagen and β‑catenin, which are phenotypic markers of the differentiation and dedifferentiation of chondrocytes. Cilostazol also significantly induced HO‑1 expression, and the induction of HO‑1 expression was affected by a significant increase in reactive oxygen species (ROS) production caused by cilostazol treatment. Of note, pre‑treatment with 3‑morpholinosydnonimine hydrochloride (SIN‑1), an inducer of HO‑1 expression, markedly attenuated cilostazol‑induced chondrocyte senescence, and thus, we examined whether HO‑1 directly modulates chondrocyte senescence induced by cilostazol. The upregulation of HO‑1 was found to suppress cilostazol‑induced cellular senescence. In addition, the inhibition of HO‑1 activity with the iron chelator, desferrioxamine (DFO), or HO‑1 siRNA increased cilostazol‑induced chondrocyte senescence. Based on these results, it can be concluded that HO‑1 is associated with the suppression of chondrocyte senescence, and that the enforced overexpression of HO‑1 protects chondrocytes against stress‑induced senescence.
Collapse
Affiliation(s)
- Kang Mi Kim
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongnam 626‑870, Republic of Korea
| | - Si Eun Park
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongnam 626‑870, Republic of Korea
| | - Mi Sun Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongnam 626‑870, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, Pusan National University School of Medicine, Yangsan, Gyeongnam 626‑870, Republic of Korea
| | - Young Chul Park
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongnam 626‑870, Republic of Korea
| |
Collapse
|
10
|
Nitric oxide compounds have different effects profiles on human articular chondrocyte metabolism. Arthritis Res Ther 2014; 15:R115. [PMID: 24025112 PMCID: PMC3978712 DOI: 10.1186/ar4295] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 06/09/2013] [Accepted: 09/11/2013] [Indexed: 01/27/2023] Open
Abstract
Introduction The pathogenesis of osteoarthritis (OA) is characterized by the production of high amounts of nitric oxide (NO), as a consequence of up-regulation of chondrocyte-inducible nitric oxide synthase (iNOS) induced by inflammatory cytokines. NO donors represent a powerful tool for studying the role of NO in the cartilage in vitro. There is no consensus about NO effects on articular cartilage in part because the differences between the NO donors available. The aim of this work is to compare the metabolic profile of traditional and new generation NO donors to see which one points out the osteoarthritic process in the best way. Methods Human healthy and OA chondrocytes were isolated from patients undergoing joint replacement surgery, and primary cultured. Cells were stimulated with NO donors (NOC-12 or SNP). NO production was evaluated by the Griess method, and apoptosis was quantified by flow cytometry. Mitochondrial function was evaluated by analysing respiratory chain enzyme complexes, citrate synthase (CS) activities by enzymatic assay, mitochondrial membrane potential (Δψm) by JC-1 using flow cytometry, and ATP levels were measured by luminescence assays. Glucose transport was measured as the uptake of 2-deoxy-[3H]glucose (2-[3H]DG). Statistical analysis was performed using the Mann-Whitney U test. Results NOC-12 liberates approximately ten times more NO2- than SNP, but the level of cell death induced was not as profound as that produced by SNP. Normal articular chondrocytes stimulated with NOC-12 had reduced activity from complexes I, III y IV, and the mitochondrial mass was increased in these cells. Deleterious effects on ΔΨm and ATP levels were more profound with SNP, and this NO donor was able to reduce 2-[3H]DG levels. Both NO donors had opposite effects on lactate release, SNP diminished the levels and NOC-12 lead to lactate accumulation. OA chondrocytes incorporate significantly more 2-[3H]DG than healthy cells. Conclusions These findings suggest that the new generation donors, specifically NOC-12, mimic the OA metabolic process much better than SNP. Previous results using SNP have to be considered prudently since most of the effects observed can be induced by the interactions of secondary products of NO.
Collapse
|
11
|
Cottier KE, Fogle EM, Fox DA, Ahmed S. Noxa in rheumatic diseases: present understanding and future impact. Rheumatology (Oxford) 2013; 53:1539-46. [PMID: 24352336 DOI: 10.1093/rheumatology/ket408] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Impaired programmed cell death is an important contributing mechanism in the development of chronic inflammatory and autoimmune diseases. Overexpression of Bcl-2 family proteins in such diseases has led to the concept of targeted suppression of these proteins as a primary therapeutic strategy. However, limited success with this approach has prompted pharmacologists to look at the other side of the coin, with the aim of reactivating jeopardized pro-apoptotic proteins that may neutralize Bcl-2 or other anti-apoptotic molecules. In this effort, BH3-only proteins have gained recent attention as endogenous molecules for the sensitization of resistant cells to undergo apoptosis. Among the BH3-only family, Noxa stands out as exceptional for its specificity to bind Mcl-1 and Bcl-2 and blunt their biological properties. Noxa is now being tested as a promising therapeutic target in cancer biology. Nonetheless, its role and clinical application still lack validation in autoimmune diseases, including rheumatic conditions. This is partly attributed to the significant gap in our understanding of its regulatory role and how either overexpression of Noxa or delivery of BH3 mimetics could be therapeutically exploited. In this review we highlight some recent studies in RA, OA, SLE and SS suggesting that Noxa may be used as a potential therapeutic target to circumvent invasive and tissue destructive processes in these rheumatic diseases.
Collapse
Affiliation(s)
- Karissa E Cottier
- Department of Pharmacology, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH and Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elise M Fogle
- Department of Pharmacology, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH and Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David A Fox
- Department of Pharmacology, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH and Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Salahuddin Ahmed
- Department of Pharmacology, College of Pharmacy & Pharmaceutical Sciences, University of Toledo, Toledo, OH and Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Mawatari T, Nakamichi I, Suenaga E, Maloney WJ, Smith RL. Effects of heme oxygenase-1 on bacterial antigen-induced articular chondrocyte catabolism in vitro. J Orthop Res 2013; 31:1943-9. [PMID: 24038461 DOI: 10.1002/jor.22394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 05/01/2013] [Indexed: 02/04/2023]
Abstract
This study tested the hypothesis that heme oxygenase-1 (HO-1) expression counteracts bacterial antigen-induced catabolic metabolism in human articular chondrocytes. HO-1 expression was induced in chondrocytes by the iron-containing porphoryin, hemin. Anti-catabolic and anti-apoptotic effects of HO-1 expression were evaluated following bacterial antigen (lipopolysaccharides, LPS) activation of chondrocytes by quantification of cytokine and cartilage matrix protein expression. Effects of HO-1 over-expression on chondrocyte matrix metabolism were evaluated using plasmid-driven protein synthesis. Hemin increased HO-1 expression and LPS increased interleukin-1beta and interleukin-6 gene and protein expression in chondrocytes. Hemin-induced HO-1 decreased LPS-induced interleukin-1beta and interleukin-6 gene and protein expression. Increased HO-1 expression partially reversed LPS-suppression of aggrecan and type II collagen gene expression and suppressed LPS-induced gene expression of IL-6, inducible nitric oxide synthase (iNOS), matrix metalloproteinases (MMPs), and IL-1beta. HO-1 induction was inversely correlated with LPS-induced chondrocyte apoptosis. HO-1 over-expression in chondrocytes decreased matrix protein gene expression. With LPS activation, increased HO-1 expression decreased chondrocyte catabolism, partially reversed LPS-dependent inhibition of cartilage matrix protein expression and protected against apoptosis. Without LPS, hemin-induced HO-1 and plasmid-based over-expression of HO-1 inhibited cartilage matrix gene expression. The results suggest that elevated HO-1 expression in chondrocytes is protective of cartilage in inflamed joints but may otherwise suppress matrix turn over.
Collapse
Affiliation(s)
- Taro Mawatari
- RR&D Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, 94304; Orthopaedic Research Laboratories, Stanford University School of Medicine, 300 Pasteur Drive, R105, Stanford, California, 94305
| | | | | | | | | |
Collapse
|
13
|
Blouquit-Laye S, Dannhoffer L, Braun C, Dinh-Xuan AT, Sage E, Chinet T. Effect of nitric oxide on epithelial ion transports in noncystic fibrosis and cystic fibrosis human proximal and distal airways. Am J Physiol Lung Cell Mol Physiol 2012; 303:L617-25. [DOI: 10.1152/ajplung.00368.2011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The airways of patients with cystic fibrosis (CF) exhibit decreased nitric oxide (NO) concentrations, which might affect airway function. The aim of this study was to determine the effects of NO on ion transport in human airway epithelia. Primary cultures of non-CF and CF bronchial and bronchiolar epithelial cells were exposed to the NO donor sodium nitroprusside (SNP), and bioelectric variables were measured in Ussing chambers. Amiloride was added to inhibit the Na+channel ENaC, and forskolin and ATP were added successively to stimulate cAMP- and Ca2+-dependent Cl−secretions, respectively. The involvement of cGMP was assessed by measuring the intracellular cGMP concentration in bronchial cells exposed to SNP and the ion transports in cultures exposed to 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one, an inhibitor of the soluble guanylate cyclase (ODQ), or to 8Z, a cocktail of 8-bromo-cGMP and zaprinast (phosphodiesterase 5 inhibitor). SNP decreased the baseline short-circuit current ( Isc) and the changes in Iscinduced by amiloride, forskolin, and ATP in non-CF bronchial and bronchiolar cultures. The mechanism of this inhibition was studied in bronchial cells. SNP increased the intracellular cGMP concentration ([cGMP]i). The inhibitory effect of SNP was abolished by 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide, an NO scavenger (PTIO) and ODQ and was partly mimicked by increasing [cGMP]i. In CF cultures, SNP did not significantly modify ion transport; in CF bronchial cells, 8Z had no effect; however, SNP increased the [cGMP]i. In conclusion, exogenous NO may reduce transepithelial Na+absorption and Cl−secretion in human non-CF airway epithelia through a cGMP-dependent pathway. In CF airways, the NO/cGMP pathway appears to exert no effect on transepithelial ion transport.
Collapse
Affiliation(s)
- Sabine Blouquit-Laye
- UPRES EA220, UFR Paris Ile de France Ouest, Université de Versailles Saint Quentin en Yvelines, Boulogne, France
| | - Luc Dannhoffer
- UPRES EA220, UFR Paris Ile de France Ouest, Université de Versailles Saint Quentin en Yvelines, Boulogne, France
| | - Camille Braun
- UPRES EA220, UFR Paris Ile de France Ouest, Université de Versailles Saint Quentin en Yvelines, Boulogne, France
| | - Anh-Tuan Dinh-Xuan
- Service de Physiologie-Explorations Fonctionnelles, Hôpital Cochin, Paris, France; and
| | - Edouard Sage
- Service de chirurgie thoracique, Hôpital Foch, Suresnes, France
| | - Thierry Chinet
- UPRES EA220, UFR Paris Ile de France Ouest, Université de Versailles Saint Quentin en Yvelines, Boulogne, France
| |
Collapse
|
14
|
Kim HA, Yeo Y, Jung HA, Jung YO, Park SJ, Kim SJ. Phase 2 enzyme inducer sulphoraphane blocks prostaglandin and nitric oxide synthesis in human articular chondrocytes and inhibits cartilage matrix degradation. Rheumatology (Oxford) 2012; 51:1006-16. [PMID: 22332123 DOI: 10.1093/rheumatology/ker525] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE We explored the inhibitory effect of sulphoraphane (SFN), a potent inducer of Phase 2 enzymes, on cytokine-induced prostaglandin E(2) (PGE2) and nitric oxide (NO) production and cartilage degradation in articular chondrocytes. The regulatory mechanism of SFN on nuclear factor (NF)-κB was investigated. METHODS Chondrocytes were obtained from patients with knee OA. Chondrocytes were stimulated with IL-1β or TNF-α with or without pre-incubation with SFN. Production of PGE2 and NO was evaluated by the Griess reaction and an ELISA. The expression of microsomal PGE synthase (mPGES), cyclo-oxygenase (COX)-2 and inducible NO synthase (iNOS) was evaluated by real-time RT-PCR and western blot analysis. The regulation of NF-κB activity was explored using luciferase and chromatin immunoprecipitation assays as well as a western blot for phosphorylated IκB kinase (IKK), IκB and the degradation of IκB. Proteoglycan and type II collagen degradation products released from explant cultures were analysed using the dimethylmethylene blue assay and an ELISA for C-terminal telopeptides of type II collagen. RESULTS SFN inhibited the production of PGE2 and NO induced by IL-1β and TNF-α. At a concentration as low as 5 μM, SFN completely inhibited mPGES, COX-2 and iNOS at the mRNA and protein levels, and proteoglycan and type II collagen degradation product release in explant culture. Various signalling pathways required for the NF-κB activation were affected by SFN. CONCLUSION SFN inhibited a broad range of catabolic mechanisms in articular chondrocytes. SFN may be a safe and effective candidate drug for the inhibition of cartilage degradation in arthritic diseases.
Collapse
Affiliation(s)
- Hyun-Ah Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyongchondong, Dongan-gu, Anyang, Kyunggi-do 431-070, Korea.
| | | | | | | | | | | |
Collapse
|
15
|
Su SH, Jen CJ, Chen HI. NO signaling in exercise training-induced anti-apoptotic effects in human neutrophils. Biochem Biophys Res Commun 2010; 405:58-63. [PMID: 21195695 DOI: 10.1016/j.bbrc.2010.12.123] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2010] [Accepted: 12/24/2010] [Indexed: 11/30/2022]
Abstract
Short-lived neutrophils play a predominant role in innate immunity, the effects of exercise training on neutrophil survival is unclear. In this study, we investigated the underlying mechanisms of training effects on human neutrophil apoptosis. Healthy male subjects were trained on a cycling ergometer for 8 weeks and followed by 4 weeks of detraining. Blood neutrophils were collected before exercise, after training, and after detraining. Comparing with pre-exercise specimens, neutrophils collected after training showed reduced apoptosis rate, which partially returned after detraining. Various intracellular proteins, including iNOS, Mcl-1, A1, Grp78, and IL-8, were upregulated by training, and they remained high after detraining. Upregulated iNOS was closely correlated with these anti-apoptotic molecules in neutrophils. Furthermore, the possible mechanism by which iNOS suppressed apoptosis was explored. Neutrophil apoptosis was accelerated by blocking and retarded by stimulating the endogenous iNOS activity. As an anti-apoptosis mediator of NO signaling, the Mcl-1 level dropped by depletion of the major NO downstream molecule cGMP and such loss of Mcl-1 was avoidable when supplying exogenous NO. Upon activation of NO-cGMP signaling, neutrophils held increased Mcl-1 expression and delayed apoptosis. Collectively, our results suggested that exercise training may retard neutrophil apoptosis by upregulating the iNOS-NO-cGMP-Mcl-1 pathway.
Collapse
Affiliation(s)
- Shu-Hui Su
- Institute of Medical Sciences, College of Medicine, Tzu Chi University, Hualien, Taiwan.
| | | | | |
Collapse
|
16
|
Gomaa A, Elshenawy M, Afifi N, Mohammed E, Thabit R. Influence of dipyridamole and its combination with NO donor or NO synthase inhibitor on adjuvant arthritis. Int Immunopharmacol 2010; 10:1406-14. [PMID: 20800711 DOI: 10.1016/j.intimp.2010.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2010] [Revised: 08/09/2010] [Accepted: 08/09/2010] [Indexed: 12/19/2022]
Abstract
The anti-arthritic and anti-inflammatory effects of dipyridamole and the possible involvement of NO in the dipyridamole action are not yet clear. The aim of this study was to evaluate the effects of dipyridamole alone and in combination with either the nitric oxide donor, sodium nitroprusside (SNP) or the non-selective nitric oxide synthase inhibitor, L-NG- monomethyl arginine (L-NMMA), on pathogenesis of adjuvant-induced arthritis model in rats. The results of the present work showed that prophylactic administration of dipyridamole alone and dipyridamole administration in combination with either low dose of SNP or L-NMMA significantly ameliorated pathogenesis of adjuvant arthritis in rats as evidenced by significant decrease in arthritis index, hind paws volume, loss of body weight, hyperalgesia compared with control vehicle (1% DMSO) treated adjuvant arthritic rats. Inflammatory cellular infiltrate in synovium of ankle joint and pannus formation were also markedly inhibited. Interleukin-10(IL-10) levels were significantly increased in these groups of animals. In contrast, a high dose of SNP counteracted the anti-inflammatory and anti-arthritic effects of dipyridamole. The inhibitory effect of therapeutic administration of dipyridamole alone on adjuvant arthritis syndrome was not significantly different from that of vehicle administration. In conclusion, dipyridamole has prophylactic but not therapeutic anti-arthritic and anti-inflammatory effects that appear to be dependent on inhibition of NO synthase. A synergistic combination between dipyridamole and NO synthase inhibitor or low dose of NO donor may have prophylactic and therapeutic values in autoimmune diseases like RA.
Collapse
Affiliation(s)
- Adel Gomaa
- Department of Pharmacology & Toxicology, College of Pharmacy, Taibah University, Al-madinah Almunawwarah, KSA.
| | | | | | | | | |
Collapse
|
17
|
Kim HA, Yeo Y, Kim WU, Kim S. Phase 2 enzyme inducer sulphoraphane blocks matrix metalloproteinase production in articular chondrocytes. Rheumatology (Oxford) 2009; 48:932-8. [PMID: 19491303 DOI: 10.1093/rheumatology/kep132] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES In addition to its chemopreventive activity, phase 2 enzyme inducers have been recently found to have anti-inflammatory activity. In this study, we examined the influence of sulphoraphane (SPN), one of the most potent inducers of the phase II enzymes on the production of MMPs by pro-inflammatory cytokines in human articular chondrocytes. METHODS Articular cartilages were obtained from knee OA patients and were cultured in monolayers and explants. Induction of a phase II enzyme, NAD(P)H:quinone oxidoreductase 1 (NQO1), in chondrocytes was assayed after incubation with various concentrations of SPN. Chondrocytes were stimulated with IL-1 or TNF-alpha with or without pre-incubation with SPN. The expression and activation of MMP-1, -3 and -13 was evaluated by an ELISA, gel zymography and RT-PCR. MAP kinases [p38, extracellular signal-regulated protein kinase (ERK) and C-Jun N terminal kinase (JNK)] and NF-kappaB activation were evaluated by western blotting and by an electrophoretic mobility shift assay, respectively. RESULTS SPN significantly induced NQO1 activity in chondrocytes and the induction was maximal at 24 h. SPN inhibited the production of MMP-1, -3 and -13 protein and mRNA induced by either IL-1 or TNF-alpha in a dose-dependent manner. This inhibition of MMP by SPN was accompanied by the inhibition of NF-kappaB and JNK activation. CONCLUSIONS SPN was found to inhibit MMP production in pro-inflammatory cytokine-stimulated chondrocytes. Delineation of the biochemical mechanism regulating cartilage catabolism by SPN may identify safe and effective therapeutic targets for the inhibition of cartilage degradation.
Collapse
Affiliation(s)
- Hyun Ah Kim
- Division of Rheumatology, Department of Internal Medicine, Hallym University Sacred Heart Hospital, Dongan-Gu, Anyang, Korea.
| | | | | | | |
Collapse
|
18
|
Dual effect of nitric oxide donor on adjuvant arthritis. Int Immunopharmacol 2009; 9:439-47. [DOI: 10.1016/j.intimp.2009.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 01/04/2009] [Accepted: 01/15/2009] [Indexed: 11/23/2022]
|
19
|
Megías J, Guillén MI, Clérigues V, Rojo AI, Cuadrado A, Castejón MA, Gomar F, Alcaraz MJ. Heme oxygenase-1 induction modulates microsomal prostaglandin E synthase-1 expression and prostaglandin E(2) production in osteoarthritic chondrocytes. Biochem Pharmacol 2009; 77:1806-13. [PMID: 19428335 DOI: 10.1016/j.bcp.2009.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2008] [Revised: 03/05/2009] [Accepted: 03/06/2009] [Indexed: 12/21/2022]
Abstract
Pro-inflammatory cytokines such as interleukin-1beta (IL-1beta) may participate in the pathogenesis of cartilage damage in osteoarthritis (OA) through the production of catabolic enzymes and inflammatory mediators. Induction of heme oxygenase-1 (HO-1) has previously been shown to exert anti-inflammatory effects in different cell types. We have investigated whether HO-1 induction may modify chondrocyte viability and the production of relevant mediators such as oxidative stress and prostaglandin E(2) (PGE(2)) elicited by IL-1beta in OA chondrocytes. Chondrocytes were isolated from OA cartilage and used in primary culture. Cells were stimulated with IL-1beta in the absence or presence of the HO-1 inducer cobalt protoporphyrin IX (CoPP). Gene expression was assessed by quantitative real-time PCR, protein levels by ELISA and Western blot, apoptosis by laser scanning cytometry using annexin V-FITC and TUNEL assays, and oxidative stress by LSC with dihydrorhodamine 123. HO-1 induction by CoPP enhanced chondrocyte viability and aggrecan content while inhibiting apoptosis and oxidative stress generation. PGE(2) is produced in OA chondrocytes stimulated by IL-1beta by the coordinated induction of cyclooxygenase-2 and microsomal PGE synthase 1 (mPGES-1). The production of PGE(2) was decreased by HO-1 induction as a result of diminished mPGES-1 protein and mRNA expression. Transfection with HO-1 small interfering RNA counteracted CoPP effects. In addition, the activation of nuclear factor-kappaB and early growth response-1 was significantly reduced by CoPP providing a basis for its anti-inflammatory effects. These results confirm the protective role of HO-1 induction in OA chondrocytes and suggest the potential interest of this strategy in degenerative joint diseases.
Collapse
Affiliation(s)
- Javier Megías
- Department of Pharmacology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Namdari S, Wei L, Moore D, Chen Q. Reduced limb length and worsened osteoarthritis in adult mice after genetic inhibition of p38 MAP kinase activity in cartilage. ACTA ACUST UNITED AC 2009; 58:3520-9. [PMID: 18975318 DOI: 10.1002/art.23999] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE MAP kinase p38 is part of an intracellular signaling pathway activated by environmental stress and inflammatory factors. Since in vitro studies show that inhibiting p38 activity leads to a reduction in the release of degenerative metalloproteinase from chondrocytes, we speculated that inactivation of p38 in vivo may be chondroprotective. To test this hypothesis, we examined the morphology of adult mice that express a dominant-negative (DN) p38 MAPK transgene in a cartilage-specific manner. METHODS The in vivo effects of the genetic inhibition of p38 MAPK activity in cartilage were investigated in 1-year-old heterozygous DN p38-transgenic mice (n = 10) using morphologic measurements, microfocal computed tomography scanning, biomechanical testing, and histologic analysis. Results were compared with those in wild-type (WT) littermates (n = 9). RESULTS Adult DN p38 MAPK+/- -transgenic mice exhibited 50% p38 MAPK activity in articular chondrocytes as compared with WT mice. They were significantly shorter in overall body length as well as in the femur and tibia lengths. There were no differences in bone material or mechanical properties between the transgenic and WT mice. Surprisingly, the transgenic mice had higher grades of osteoarthritis of the knee joint. CONCLUSION Genetic inhibition of p38 MAPK activity in cartilage results in shortened limb length and defects in the articular cartilage of the knee joints of adult mice. Our findings demonstrate that chronic life-long reduction of p38 MAPK activity may be harmful to joint health and suggest that the timing of p38 inhibition for chondroprotection in vivo is an important variable that warrants further investigation.
Collapse
Affiliation(s)
- Surena Namdari
- Rhode Island Hospital, and Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
| | | | | | | |
Collapse
|
21
|
Guillén M, Megías J, Gomar F, Alcaraz M. Haem oxygenase-1 regulates catabolic and anabolic processes in osteoarthritic chondrocytes. J Pathol 2008; 214:515-22. [PMID: 18200630 DOI: 10.1002/path.2313] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Pro-inflammatory cytokines, matrix metalloproteinases (MMPs) and other catabolic factors participate in the pathogenesis of cartilage damage in osteoarthritis (OA). Pro-inflammatory cytokines such as interleukin-1beta (IL-1beta) mediate cartilage degradation and might be involved in the progression of OA. Previously, we found that haem oxygenase-1 (HO-1) is down-regulated by pro-inflammatory cytokines and up-regulated by IL-10 in OA chondrocytes. The aim of this study was to determine whether HO-1 can modify the catabolic effects of IL-1beta in OA cartilage and chondrocytes. Up-regulation of HO-1 by cobalt protoporphyrin IX significantly reduced glycosaminoglycan degradation elicited by IL-1beta in OA cartilage explants but increased glycosaminoglycan synthesis and the expression of collagen II in OA chondrocytes in primary culture, as determined by radiometric procedures, immunoblotting and immunocytochemistry. HO-1 decreased the activation of extracellular signal-regulated kinase 1/2. This was accompanied by a significant inhibition in MMP activity and expression of collagenases MMP-1 and MMP-13 at the protein and mRNA levels. In addition, HO-1 induction caused a significant increase in the production of insulin-like growth factor-1 and a reduction in the levels of insulin-like growth factor binding protein-3. We have shown in primary culture of chondrocytes and articular explants from OA patients that HO-1 counteracts the catabolic and anti-anabolic effects of IL-1beta. Our data thus suggest that HO-1 may be a factor regulating the degradation and synthesis of extracellular matrix components in OA.
Collapse
Affiliation(s)
- Mi Guillén
- Department of Pharmacology, Faculty of Pharmacy, University of Valencia, Spain
| | | | | | | |
Collapse
|
22
|
Yoo SA, Park BH, Yoon HJ, Lee JY, Song JH, Kim HA, Cho CS, Kim WU. Calcineurin modulates the catabolic and anabolic activity of chondrocytes and participates in the progression of experimental osteoarthritis. ACTA ACUST UNITED AC 2007; 56:2299-311. [PMID: 17599750 DOI: 10.1002/art.22731] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To determine whether intracellular calcineurin (Cn), a calcium-activated phosphatase, regulates the anabolic and catabolic activities of chondrocytes, and is a potential target in the treatment of osteoarthritis (OA). METHODS CnA expression was examined in cartilage tissue samples and cultured chondrocytes from OA patients, using immunohistochemistry and Western blot analysis, respectively. Concentrations of matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases 1 (TIMP-1) in the culture supernatants were determined using enzyme-linked immunosorbent assay. Levels of nitric oxide (NO) and type II collagen (CII) were measured using the Griess reaction and Western blot analysis, respectively. In addition, the pathologic role of Cn was examined in an in vivo model in which experimental OA was induced in mice by injecting type VII collagenase into the knee joints. RESULTS CnA was highly expressed in the chondrocytes of lesional OA cartilage. Cyclosporin A (CSA), a Cn inhibitor, inhibited spontaneous and interleukin-1beta-stimulated production of NO, MMP-1, and MMP-3 in chondrocytes. However, CSA increased the levels of production of CII, TIMP-1, and transforming growth factor beta. Similar changes in MMP-1, NO, and CII expression levels in chondrocytes were observed after the targeted inhibition of Cn by overexpression of calcineurin binding protein 1, a natural Cn antagonist. Moreover, in the mouse model, animals treated with CSA showed a significant decrease in both the extent and the severity of cartilage damage, which were assessed macroscopically and microscopically, compared with vehicle-treated animals. CONCLUSION These results suggest that CnA is critically involved in the catabolic and anabolic activities of chondrocytes as well as in the progression of experimental OA. Targeted inhibition of CnA may be an effective treatment strategy for OA.
Collapse
Affiliation(s)
- Seung-Ah Yoo
- Catholic University of Korea, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Kim HA, Won Kim D, Park J, Choi SY. Transduction of Cu, Zn-superoxide dismutase mediated by an HIV-1 Tat protein basic domain into human chondrocytes. Arthritis Res Ther 2007; 8:R96. [PMID: 16792821 PMCID: PMC1779403 DOI: 10.1186/ar1972] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 05/04/2006] [Accepted: 05/12/2006] [Indexed: 01/01/2023] Open
Abstract
This study was performed to investigate the transduction of a full-length superoxide dismutase (SOD) protein fused to transactivator of transcription (Tat) into human chondrocytes, and to determine the regulatory function of transduced Tat-SOD in the inflammatory cytokine induced catabolic pathway. The pTat-SOD expression vector was constructed to express the basic domain of HIV-1 Tat as a fusion protein with Cu, Zn-SOD. We also purified histidine-tagged SOD without an HIV-1 Tat and Tat-GFP as control proteins. Cartilage samples were obtained from patients with osteoarthritis (OA) and chondrocytes were cultured in both a monolayer and an explant. For the transduction of fusion proteins, cells/explants were treated with a variety of concentrations of fusion proteins. The transduced protein was detected by fluorescein labeling, western blotting and SOD activity assay. Effects of transduced Tat-SOD on the regulation of IL-1 induced nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) mRNA expression was assessed by the Griess reaction and reverse transcriptase PCR, respectively. Tat-SOD was successfully delivered into both the monolayer and explant cultured chondrocytes, whereas the control SOD was not. The intracellular transduction of Tat-SOD into cultured chondrocytes was detected after 1 hours, and the amount of transduced protein did not change significantly after further incubation. SOD enzyme activity increased in a dose-dependent manner. NO production and iNOS mRNA expression, in response to IL-1 stimulation, was significantly down-regulated by pretreatment with Tat-SOD fusion proteins. This study shows that protein delivery employing the Tat-protein transduction domain is feasible as a therapeutic modality to regulate catabolic processes in cartilage. Construction of additional Tat-fusion proteins that can regulate cartilage metabolism favorably and application of this technology in in vivo models of arthritis are the subjects of future studies.
Collapse
Affiliation(s)
- Hyun Ah Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyongchondong, Dongan-gu, Anyang, Kyunggi-do, 431-070, Korea
| | - Dae Won Kim
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Jinseu Park
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Soo Young Choi
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| |
Collapse
|
24
|
Kwak HJ, Park KM, Lee S, Lim HJ, Go SH, Eom SM, Park HY. Preconditioning with low concentration NO attenuates subsequent NO-induced apoptosis in vascular smooth muscle cells via HO-1-dependent mitochondrial death pathway. Toxicol Appl Pharmacol 2006; 217:176-84. [PMID: 17027882 DOI: 10.1016/j.taap.2006.08.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2006] [Revised: 07/28/2006] [Accepted: 08/28/2006] [Indexed: 11/16/2022]
Abstract
Nitric oxide (NO) signaling pathways are important in both the maintenance of vascular homeostasis and disease progression. Overproduction of NO has been associated with ischemia/reperfusion (I/R) injury. Growing evidences suggest that NO preconditioning has cytoprotective effects against I/R injury. However, the mechanism with which NO mediates these effects remains to be elucidated. The purpose of this study was to examine the mechanism of how NO preconditioning inhibits subsequent NO-induced apoptosis in vascular smooth muscle cells (VSMC), specifically focusing on heme oxygenase-1 (HO-1). According to our data, sodium nitroprusside (SNP) increased HO-1 expression in a concentration dependent manner. Preconditioning with low concentration SNP (0.3mM) inhibited subsequent high concentration SNP (1.5mM)-induced apoptosis, and this effect was reversed by the HO-1 inhibitor SnPP. Low concentration SNP-mediated protection involved p38 kinase inactivation and increased Bcl-2 expression. Furthermore, mitochondrial membrane potential was concomitantly increased with decreased expressions of Bax, Apaf-1, and activity of caspase-3, which was reversed by SnPP treatment. Our results show that low concentration SNP preconditioning suppresses subsequent high concentration SNP-induced apoptosis by inhibiting p38 kinase and mitochondrial death pathway via HO-1-dependent mechanisms in VSMC.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Apoptosis/drug effects
- Apoptosis Regulatory Proteins/metabolism
- Cell Survival/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Heme Oxygenase (Decyclizing)/metabolism
- Male
- Membrane Potential, Mitochondrial/drug effects
- Mitochondria/drug effects
- Mitochondria/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Nitric Oxide/metabolism
- Nitric Oxide Donors/pharmacology
- Nitroprusside/pharmacology
- Phosphorylation
- Rats
- Rats, Wistar
- Up-Regulation/drug effects
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Hyun-Jeong Kwak
- Division of Cardiovascular Diseases, Center for Biomedical Sciences, National Institutes of Health, Nokbun-dong, Eunpyung-gu, Seoul 122-701, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|