51
|
Antarctic Krill Oil improves articular cartilage degeneration via activating chondrocyte autophagy and inhibiting apoptosis in osteoarthritis mice. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
52
|
Meng Z, Shen B, Gu Y, Wu Z, Yao J, Bian Y, Zeng D, Chen K, Cheng S, Fu J, Peng L, Zhao Y. Diazoxide ameliorates severity of experimental osteoarthritis by activating autophagy via modulation of the osteoarthritis-related biomarkers. J Cell Biochem 2018; 119:8922-8936. [PMID: 29953665 DOI: 10.1002/jcb.27145] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/18/2018] [Indexed: 01/05/2023]
Abstract
Accumulating evidence suggests that autophagy plays a protective role in chondrocytes and prevents cartilage degeneration in osteoarthritis (OA). The objective of this study was to investigate the effect of diazoxide on chondrocyte death and cartilage degeneration and to determine whether these effects are correlated to autophagy in experimental OA. In this study, a cellular OA model was established by stimulating SW1353 cells with interleukin 1β. A rat OA model was generated by transecting the anterior cruciate ligament combined with the resection of the medial menisci, followed by treatment with diazoxide or diazoxide combination with 3-methyladenine. The percentage of viable cells was evaluated using calcein-acetoxymethyl/propidium iodide double staining. The messenger RNA expression levels of collagen type II alpha 1 chain (COL2A1), matrix metalloproteinase 13 (MMP-13), TIMP metallopeptidase inhibitor 1 (TIMP-1), and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) were determined using quantitative real-time polymerase chain reaction. The cartilage thickness and joint space were evaluated using ultrasound. SW1353 cell degeneration and autophagosomes were observed using transmission electron microscopy. The expression levels of microtubule-associated protein 1 light chain 3 (LC3), beclin-1, P62, COL2A1, and MMP-13 were evaluated using immunofluorescence staining and Western blot analysis. Diazoxide significantly attenuated articular cartilage degeneration and SW1353 cell death in experimental OA. The restoration of autophagy was observed in the diazoxide-treated group. The beneficial effects of diazoxide were markedly blocked by 3-methyladenine. Diazoxide treatment also modulated the expression levels of OA-related biomarkers. These results demonstrated that diazoxide exerted a chondroprotective effect and attenuated cartilage degeneration by restoring autophagy via modulation of OA-related biomarkers in experimental OA. Diazoxide treatment might be a promising therapeutic approach to prevent the development of OA.
Collapse
Affiliation(s)
- ZhuLong Meng
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - BiXin Shen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - YunTao Gu
- Department of Orthopeadic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - ZiQuan Wu
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - JiangLing Yao
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - YangYang Bian
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - DeLu Zeng
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - KeWei Chen
- Department of Orthopeadic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - ShaoWen Cheng
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Jian Fu
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Lei Peng
- Department of Trauma Center, The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - YingZheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
53
|
Vinatier C, Domínguez E, Guicheux J, Caramés B. Role of the Inflammation-Autophagy-Senescence Integrative Network in Osteoarthritis. Front Physiol 2018; 9:706. [PMID: 29988615 PMCID: PMC6026810 DOI: 10.3389/fphys.2018.00706] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis is the most common musculoskeletal disease causing chronic disability in adults. Studying cartilage aging, chondrocyte senescence, inflammation, and autophagy mechanisms have identified promising targets and pathways with clinical translatability potential. In this review, we highlight the most recent mechanistic and therapeutic preclinical models of aging with particular relevance in the context of articular cartilage and OA. Evidence supporting the role of metabolism, nuclear receptors and transcription factors, cell senescence, and circadian rhythms in the development of musculoskeletal system degeneration assure further translational efforts. This information might be useful not only to propose hypothesis and advanced models to study the molecular mechanisms underlying joint degeneration, but also to translate our knowledge into novel disease-modifying therapies for OA.
Collapse
Affiliation(s)
- Claire Vinatier
- INSERM, UMR 1229, Regenerative Medicine and Skeleton, University of Nantes, ONIRIS, Nantes, France.,University of Nantes, UFR Odontologie, Nantes, France
| | - Eduardo Domínguez
- Biofarma Research Group, Center for Research in Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jerome Guicheux
- INSERM, UMR 1229, Regenerative Medicine and Skeleton, University of Nantes, ONIRIS, Nantes, France.,University of Nantes, UFR Odontologie, Nantes, France.,CHU Nantes, PHU4 OTONN, Nantes, France
| | - Beatriz Caramés
- Grupo de Biología del Cartílago, Servicio de Reumatología. Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, Sergas, A Coruña, Spain
| |
Collapse
|
54
|
Chen LY, Lotz M, Terkeltaub R, Liu-Bryan R. Modulation of matrix metabolism by ATP-citrate lyase in articular chondrocytes. J Biol Chem 2018; 293:12259-12270. [PMID: 29929979 DOI: 10.1074/jbc.ra118.002261] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 06/04/2018] [Indexed: 02/04/2023] Open
Abstract
Certain dysregulated chondrocyte metabolic adaptive responses such as decreased activity of the master regulator of energy metabolism AMP-activated protein kinase (AMPK) promote osteoarthritis (OA). Metabolism intersects with epigenetic and transcriptional responses. Hence, we studied chondrocyte ATP-citrate lyase (ACLY), which generates acetyl-CoA from mitochondrial-derived citrate, and modulates acetylation of histones and transcription factors. We assessed ACLY in normal and OA human knee chondrocytes and cartilages by Western blotting and immunohistochemistry, and quantified acetyl-CoA fluorometrically. We examined histone and transcription factor lysine acetylation by Western blotting, and assessed histone H3K9 and H3K27 occupancy of iNOS, MMP3, and MMP13 promoters by chromatin immunoprecipitation (ChIP) and quantitative PCR (qPCR). We analyzed iNOS, MMP3, MMP13, aggrecan (ACAN), and Col2a1 gene expression by RT-qPCR. Glucose availability regulated ACLY expression and function, nucleocytosolic acetyl-CoA, and histone acetylation. Human knee OA chondrocytes exhibited increased ACLY activation (assessed by Ser-455 phosphorylation), associated with increased H3K9 and H3K27 acetylation. Inhibition of ACLY attenuated IL-1β-induced transcription of iNOS, MMP3, and MMP13 by suppressing acetylation of p65 NF-κB, H3K9, and H3K27, blunted release of NO, MMP3, and MMP13, and also reduced SOX9 acetylation that promoted SOX9 nuclear translocation, leading to increased aggrecan and Col2a1 mRNA expression. ACLY is a novel player involved in regulation of cartilage matrix metabolism. Increased ACLY activity in OA chondrocytes increased nucleocytosolic acetyl-CoA, leading to increased matrix catabolism via dysregulated histone and transcription factor acetylation. Pharmacologic ACLY inhibition in OA chondrocytes globally reverses these changes and stimulates matrix gene expression and AMPK activation, supporting translational investigation in OA.
Collapse
Affiliation(s)
- Liang-Yu Chen
- Veterans Affairs San Diego Healthcare System, San Diego, California 92161
| | - Martin Lotz
- The Scripps Research Institute, La Jolla, California 92037
| | - Robert Terkeltaub
- Veterans Affairs San Diego Healthcare System, San Diego, California 92161; Department of Medicine, University of California San Diego, La Jolla, California 92037
| | - Ru Liu-Bryan
- Veterans Affairs San Diego Healthcare System, San Diego, California 92161; Department of Medicine, University of California San Diego, La Jolla, California 92037.
| |
Collapse
|
55
|
Sun H, Wu Y, Pan Z, Yu D, Chen P, Zhang X, Wu H, Zhang X, An C, Chen Y, Qin T, Lei X, Yuan C, Zhang S, Zou W, Ouyang H. Gefitinib for Epidermal Growth Factor Receptor Activated Osteoarthritis Subpopulation Treatment. EBioMedicine 2018; 32:223-233. [PMID: 29898872 PMCID: PMC6020860 DOI: 10.1016/j.ebiom.2018.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/07/2018] [Accepted: 06/04/2018] [Indexed: 01/18/2023] Open
Abstract
Osteoarthritis (OA) is a leading cause of physical disability among aging populations, with no available drugs able to efficiently restore the balance between cartilage matrix synthesis and degradation. Also, OA has not been accurately classified into subpopulations, hindering the development toward personalized precision medicine. In the present study, we identified a subpopulation of OA patients displaying high activation level of epidermal growth factor receptor (EGFR). With Col2a1-creERT2; Egfrf/f mice, it was found that the activation of EGFR, indicated by EGFR phosphorylation (pEGFR), led to the destruction of joints. Excitingly, EGFR inhibition prohibited cartilage matrix degeneration and promoted cartilage regeneration. The Food and Drug Administration (FDA)-approved drug gefitinib could efficiently inhibit EGFR functions in OA joints and restore cartilage structure and function in the mouse model as well as the clinical case report. Overall, our findings suggested the concept of the EGFR activated OA subpopulation and illustrated the mechanism of EGFR signaling in regulating cartilage homeostasis. Gefitinib could be a promising disease-modifying drug for this OA subpopulation treatment.
Collapse
Affiliation(s)
- Heng Sun
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Yan Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Orthopeadics, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Zongyou Pan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Dongsheng Yu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Orthopedics, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Pengfei Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Xiaoan Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Haoyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Xiaolei Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Chengrui An
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Yishan Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Tian Qin
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Xiaoyue Lei
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Chunhui Yuan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Shufang Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang 310058, China; Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China; China Orthopedic Regenerative Medicine Group, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
56
|
Chen CH, Ho ML, Chang LH, Kang L, Lin YS, Lin SY, Wu SC, Chang JK. Parathyroid hormone-(1–34) ameliorated knee osteoarthritis in rats via autophagy. J Appl Physiol (1985) 2018; 124:1177-1185. [DOI: 10.1152/japplphysiol.00871.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anterior cruciate ligament (ACL) tear can lead to osteoarthritis (OA). However, parathyroid hormone (PTH)-(1–34) was found to alleviate OA progression in a papain-induced OA model. Autophagy is a protective mechanism in normal cartilage, and its aging-related loss is linked with chondrocyte death and OA. Thus we examined the roles of autophagy in PTH treatment in OA after ACL transection (ACLT). Thirty-six rats were randomized into three groups: control group, ACLT-induced OA (OA) group, and OA with intra-articular PTH-(1–34) treatment (OA+PTH) group. Weight-bearing and treadmill tests were evaluated. Cartilage matrix was determined by a histological evaluation of glycosaminoglycan (GAG), Osteoarthritis Research Society International (OARSI) score, chondrocyte apoptosis, and immunohistochemistry. Rats in the OA group had significantly decreased weight bearing and running endurance. The histological results indicated that GAG, collagen type II, and chondrocyte autophagy had decreased but that the OARSI score, terminal differentiation markers (collagen type X and Indian hedgehog), and chondrocyte apoptosis had increased in the OA group. Additionally, PTH-(1–34) treatment significantly improved weight bearing and treadmill endurance, preserved GAG and collagen type II, and reduced the OARSI score and terminal differentiation markers. Finally, PTH-(1–34) ameliorated chondrocyte apoptosis by regulating the expression of autophagy-related proteins, through reducing mechanistic target of rapamycin (mTOR) and p62 and enhancing microtubule-associated protein-1 light chain 3 (LC3) and beclin-1. Reconstructive surgery after ACL rupture cannot prevent OA occurrence. Intra-articular PTH-(1–34) treatment can alleviate OA progression after ACLT and histological molecular changes. Possible mechanisms are reducing chondrocyte terminal differentiation and apoptosis, with increasing autophagy. NEW & NOTEWORTHY Anterior cruciate ligament (ACL) tear can lead to osteoarthritis (OA). Intra-articular parathyroid hormone (PTH)-(1–34) significantly improved weight bearing and treadmill endurance, preserved glycosaminoglycan and collagen type II, and reduced Osteoarthritis Research Society International (OARSI) score and terminal differentiation. Finally, PTH-(1–34) ameliorated chondrocyte apoptosis by regulating the expression of autophagy-related proteins, through reducing mechanistic target of rapamycin (mTOR) and p62 and enhancing microtubule-associated protein-1 light chain 3 (LC3) and beclin-1. PTH-(1–34) can alleviate OA progression after ACL transection. Possible mechanisms are reducing chondrocyte terminal differentiation and apoptosis, with increasing autophagy.
Collapse
Affiliation(s)
- Chung-Hwan Chen
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ling-Hua Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lin Kang
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Shan Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sung-Yen Lin
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shun-Cheng Wu
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopaedics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Adult Reconstruction Surgery, Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
57
|
Mantripragada VP, Piuzzi NS, Zachos T, Obuchowski NA, Muschler GF, Midura RJ. High occurrence of osteoarthritic histopathological features unaccounted for by traditional scoring systems in lateral femoral condyles from total knee arthroplasty patients with varus alignment. Acta Orthop 2018; 89:197-203. [PMID: 29119853 PMCID: PMC5901518 DOI: 10.1080/17453674.2017.1398559] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background and purpose - A better understanding of the patterns and variation in initiation and progression of osteoarthritis (OA) in the knee may influence the design of therapies to prevent or slow disease progression. By studying cartilage from the human lateral femoral condyle (LFC), we aimed to: (1) assess specimen distribution into early, mild, moderate, and severe OA as per the established histopathological scoring systems (HHGS and OARSI); and (2) evaluate whether these 2 scoring systems provide sufficient tools for characterizing all the features and variation in patterns of OA. Patients and methods - 2 LFC osteochondral specimens (4 x 4 x 8 mm) were collected from 50 patients with idiopathic OA varus knee and radiographically preserved lateral compartment joint space undergoing total knee arthroplasty. These were fixed, sectioned, and stained with HE and Safranin O-Fast Green (SafO). Results - The histopathological OA severity distribution of the 100 specimens was: 6 early, 62 mild, 30 moderate, and 2 severe. Overall, 45/100 specimens were successfully scored by both HHGS and OARSI: 12 displayed low OA score and 33 displayed cartilage surface changes associated with other histopathological features. However, 55/100 samples exhibited low surface structure scores, but were deemed to be inadequately scored by HHGS and OARSI because of anomalous features in the deeper zones not accounted for by these systems: 27 exhibited both SafO and tidemark abnormal features, 16 exhibited only SafO abnormal features, and 12 exhibited tidemark abnormal features. Interpretation - LFC specimens were scored as mild to moderate OA by HHGS and OARSI. Yet, several specimens exhibited deep zone anomalies while maintaining good surface structure, inconsistent with mild OA. Overall, a better classification of these anomalous histopathological features could help better understand idiopathic OA and potentially recognize different subgroups of disease.
Collapse
Affiliation(s)
- Venkata P Mantripragada
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| | - Nicolas S Piuzzi
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, USA,Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, USA, Instituto Universitario del Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Terri Zachos
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, USA
| | - Nancy A Obuchowski
- Department of Quantitative Health Science, Cleveland Clinic, Cleveland, USA
| | - George F Muschler
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, USA,Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, USA,Correspondence:
| | - Ronald J Midura
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| |
Collapse
|
58
|
Wang ZJ, Zhang HB, Chen C, Huang H, Liang JX. Effect of PPARG on AGEs-induced AKT/MTOR signaling-associated human chondrocytes autophagy. Cell Biol Int 2018; 42:841-848. [PMID: 29453775 DOI: 10.1002/cbin.10951] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/11/2018] [Indexed: 12/21/2022]
Abstract
Accumulation of advanced glycation end products (AGEs) in articular cartilage is thought to represent a major risk factor for osteoarthritis development. In this study we aimed to probe the role of AGEs in human chondrocytes and to determine the impact of the peroxisome proliferator-activated receptor-γ (PPARG) on AGEs-induced cell autophagy. Cell viability was measured after human chondrocytes were treated with different concentrations of AGEs with or without the PPARG inhibitor, T0070907, or agonist, pioglitazone. Autophagy activation markers (MAP2LC3, BECN1 and SQSTM1/P62), expression of PPARG and the phosphorylation levels of Akt/MTOR were determined by Western blotting; autophagosome formation was analyzed by transmission electron microscopy (TEM); autophagic flux was detected with mRFP-GFP-LC3 tandem construct. Low doses of AGEs over a short amount of time stimulated chondrocyte proliferation and autophagy by limiting phosphorylation of Akt/MTOR signaling. The addition of PPARG inhibitor T0070907 lead to defective autophagy. High dose and long exposure to AGEs inhibited cell viability and autophagy by increasing phosphorylation levels of Akt/MTOR signaling. The agonist, pioglitazone, was shown to protect cell autophagy in a dose-dependent manner. Our findings suggest AGEs can downregulate PPARG and that PPARG maintains cell viability by activating the Akt/MTOR signaling pathway as well as inducing chondrocyte autophagy.
Collapse
Affiliation(s)
- Zhao-Jun Wang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 163rd Central Hospital of the People's Liberation Army, Changsha, Hunan, 410003, People's Republic of China
| | - Hai-Bin Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 163rd Central Hospital of the People's Liberation Army, Changsha, Hunan, 410003, People's Republic of China
| | - Cheng Chen
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 163rd Central Hospital of the People's Liberation Army, Changsha, Hunan, 410003, People's Republic of China
| | - Hao Huang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 163rd Central Hospital of the People's Liberation Army, Changsha, Hunan, 410003, People's Republic of China
| | - Jian-Xia Liang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 163rd Central Hospital of the People's Liberation Army, Changsha, Hunan, 410003, People's Republic of China
| |
Collapse
|
59
|
Ren P, Niu H, Gong H, Zhang R, Fan Y. Morphological, biochemical and mechanical properties of articular cartilage and subchondral bone in rat tibial plateau are age related. J Anat 2018; 232:457-471. [PMID: 29266211 PMCID: PMC5807934 DOI: 10.1111/joa.12756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2017] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to investigate age-related changes in the morphological, biochemical and mechanical properties of articular cartilage (AC) and subchondral bone in the rat tibial plateau. Female Wistar rats were grouped according to age (1, 3, 5, 7, 9, 11, 13, 15, 16 and 17 months, with 10 rats in each group). The ultrastructures, surface topographies, and biochemical and mechanical properties of the AC and subchondral bone in the knee joints of the rats were determined through X-ray micro-tomography, histology, immunohistochemistry, scanning electron microscopy (SEM), atomic force microscopy and nanoindentation. We found that cartilage thickness decreased with age. This decrease was accompanied by functional condensation of the underlying subchondral bone. Increased thickness and bone mineral density and decreased porosity were observed in the subchondral plate (SP). Growth decreased collagen II expression in the tibial cartilage. The arrangement of trabeculae in the subchondral trabecular bone became disordered. The thickness and strength of the fibers decreased with age, as detected by SEM. The SP and trabeculae in the tibial plateau increased in roughness in the first phase (1-9 months of age), and then were constant in the second phase (11-17 months of age). Meanwhile, the roughness of the AC changed significantly in the first phase (1-9 months of age), but the changes were independent of age thereafter. This study gives a comprehensive insight into the growth-related structural, biochemical and mechanical changes in the AC and subchondral bone. The results presented herein may contribute to a new understanding of the pathogenesis of age-related bone diseases.
Collapse
Affiliation(s)
- Pengling Ren
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Haijun Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Rui Zhang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of EducationSchool of Biological Science and Medical EngineeringBeihang UniversityBeijingChina
- National Research Center for Rehabilitation Technical AidsBeijingChina
| |
Collapse
|
60
|
Abstract
Osteoarthritis is characterized by a chronic, progressive and irreversible degradation of the articular cartilage associated with joint inflammation and a reparative bone response. More than 100 million people are affected by this condition worldwide with significant health and welfare costs. Our available treatment options in osteoarthritis are extremely limited. Chondral or osteochondral grafts have shown some promising results but joint replacement surgery is by far the most common therapeutic approach. The difficulty lies on the limited regeneration capacity of the articular cartilage, poor blood supply and the paucity of resident progenitor stem cells. In addition, our poor understanding of the molecular signalling pathways involved in the senescence and apoptosis of chondrocytes is a major factor restricting further progress in the area. This review focuses on molecules and approaches that can be implemented to delay or even rescue chondrocyte apoptosis. Ways of modulating the physiologic response to trauma preventing chondrocyte death are proposed. The use of several cytokines, growth factors and advances made in altering several of the degenerative genetic pathways involved in chondrocyte apoptosis and degradation are also presented. The suggested approaches can help clinicians to improve cartilage tissue regeneration.
Collapse
Affiliation(s)
- Ippokratis Pountos
- Academic Department of Trauma & Orthopaedics, School of Medicine, University of Leeds, UK.
| | - Peter V Giannoudis
- Academic Department of Trauma & Orthopaedics, School of Medicine, University of Leeds, UK; NIHR Leeds Biomedical Research Center, Chapel Allerton Hospital, Leeds, UK.
| |
Collapse
|
61
|
A standardized extract of Butea monosperma (Lam.) flowers suppresses the IL-1β-induced expression of IL-6 and matrix-metalloproteases by activating autophagy in human osteoarthritis chondrocytes. Biomed Pharmacother 2017; 96:198-207. [DOI: 10.1016/j.biopha.2017.09.140] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/12/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023] Open
|
62
|
Abstract
Osteoarthritis is characterized by continuous degeneration of articular cartilage resulting in disability. The death of chondrocytes and the loss of the extracellular matrix are the central peculiarities in cartilage degeneration during osteoarthritis pathogenesis. Autophagy is an essential cellular homeostasis mechanism whereby cellular organelles and macromolecules are recycled to maintain cellular metabolism. Autophagy is reported to be cytoprotective effects for articular cartilage, and osteoarthritis is associated with decreased autophagy. While autophagy is known to be cytoprotective to chondrocytes, its role may vary with differing stages and models of osteoarthritis. Therefore, more in-depth studies on autophagy are needed to determine its impact on cell survival and death in articular cartilage under various in vitro and in vivo conditions. Application of autophagy on osteoarthritis therapeutics will be possible after a profound understanding is established on the role of autophagy in osteoarthritis pathogenesis.
Collapse
Affiliation(s)
- Hyelin Jeon
- a Department of Orthopaedics , Dongguk University Ilsan Hospital , Goyang , Republic of Korea
| | - Gun-Il Im
- a Department of Orthopaedics , Dongguk University Ilsan Hospital , Goyang , Republic of Korea
| |
Collapse
|
63
|
Meckes JK, Caramés B, Olmer M, Kiosses WB, Grogan SP, Lotz MK, D'Lima DD. Compromised autophagy precedes meniscus degeneration and cartilage damage in mice. Osteoarthritis Cartilage 2017; 25:1880-1889. [PMID: 28801209 PMCID: PMC5650923 DOI: 10.1016/j.joca.2017.07.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 07/18/2017] [Accepted: 07/31/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Autophagy is a cellular homeostasis mechanism that facilitates normal cell function and survival. Objectives of this study were to determine associations between autophagic responses with meniscus injury, joint aging, and osteoarthritis (OA), and to establish the temporal relationship with structural changes in menisci and cartilage. METHODS Constitutive activation of autophagy during aging was measured in GFP-LC3 transgenic reporter mice between 6 and 30 months. Meniscus injury was created by surgically destabilizing the medial meniscus (DMM) to induce posttraumatic OA in C57BL/6J mice. Levels of autophagy proteins and activation were analyzed by confocal microscopy and immunohistochemistry. Associated histopathological changes, such as cellularity, matrix staining, and structural damage, were graded in the meniscus and compared to changes in articular cartilage. RESULTS In C57BL/6J mice, basal autophagy was lower in the meniscus than in articular cartilage. With increasing age, expression of the autophagy proteins ATG5 and LC3 was significantly reduced by 24 months. Age-related changes included abnormal Safranin-O staining and reduced cellularity, which preceded structural damage in the meniscus and articular cartilage. In mice with DMM, autophagy was induced in the meniscus while it was suppressed in cartilage. Articular cartilage exhibited the most profound changes in autophagy and structure that preceded meniscus degeneration. Systemic administration of rapamycin to mice with DMM induced autophagy activation in cartilage and reduced degenerative changes in both meniscus and cartilage. CONCLUSION Autophagy is significantly affected in the meniscus during aging and injury and precedes structural damage. Maintenance of autophagic activity appears critical for meniscus and cartilage integrity.
Collapse
MESH Headings
- Aging/metabolism
- Animals
- Autophagy/drug effects
- Autophagy/physiology
- Autophagy-Related Protein 5/metabolism
- Cartilage, Articular/drug effects
- Cartilage, Articular/pathology
- Green Fluorescent Proteins/genetics
- Immunosuppressive Agents/pharmacology
- Menisci, Tibial/pathology
- Menisci, Tibial/surgery
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Confocal
- Microtubule-Associated Proteins/metabolism
- Osteoarthritis, Knee/etiology
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/physiopathology
- Sirolimus/pharmacology
- Tibial Meniscus Injuries/complications
Collapse
Affiliation(s)
- J K Meckes
- Materials Science and Engineering Program, Department of Mechanical and Aerospace Engineering, University of California, San Diego, CA, USA.
| | - B Caramés
- Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, SERGAS, and Universidade da Coruña, A Coruña, Spain.
| | - M Olmer
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| | - W B Kiosses
- Core Microscopy, The Scripps Research Institute, La Jolla, CA, USA.
| | - S P Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA.
| | - M K Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| | - D D D'Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA.
| |
Collapse
|
64
|
Kang X, Yang W, Feng D, Jin X, Ma Z, Qian Z, Xie T, Li H, Liu J, Wang R, Li F, Li D, Sun H, Wu S. Cartilage-Specific Autophagy Deficiency Promotes ER Stress and Impairs Chondrogenesis in PERK-ATF4-CHOP-Dependent Manner. J Bone Miner Res 2017; 32:2128-2141. [PMID: 28304100 DOI: 10.1002/jbmr.3134] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 01/07/2023]
Abstract
Autophagy is activated during nutritionally depleted or hypoxic conditions to facilitate cell survival. Because growth plate is an avascular and hypoxic tissue, autophagy may have a crucial role during chondrogenesis; however, the functional role and underlying mechanism of autophagy in regulation of growth plate remains elusive. In this study, we generated TamCart Atg7-/- (Atg7cKO) mice to explore the role of autophagy during endochondral ossification. Atg7cKO mice exhibited growth retardation associated with reduced chondrocyte proliferation and differentiation, and increased chondrocyte apoptosis. Meanwhile, we observed that Atg7 ablation mainly induced the PERK-ATF4-CHOP axis of the endoplasmic reticulum (ER) stress response in growth plate chondrocytes. Although Atg7 ablation induced ER stress in growth plate chondrocytes, the addition of phenylbutyric acid (PBA), a chemical chaperone known to attenuate ER stress, partly neutralized such effects of Atg7 ablation on longitudinal bone growth, indicating the causative interaction between autophagy and ER stress in growth plate. Consistent with these findings in vivo, we also observed that Atg7 ablation in cultured chondrocytes resulted in defective autophagy, elevated ER stress, decreased chondrocytes proliferation, impaired expression of col10a1, MMP-13, and VEGFA for chondrocyte differentiation, and increased chondrocyte apoptosis, while such effects were partly nullified by reduction of ER stress with PBA. In addition, Atg7 ablation-mediated impaired chondrocyte function (chondrocyte proliferation, differentiation, and apoptosis) was partly reversed in CHOP-/- cells, indicating the causative role of the PERK-ATF4-CHOP axis of the ER stress response in the action of autophagy deficiency in chondrocytes. In conclusion, our findings indicate that autophagy deficiency may trigger ER stress in growth plate chondrocytes and contribute to growth retardation, thus implicating autophagy as an important regulator during chondrogenesis and providing new insights into the clinical potential of autophagy in cartilage homeostasis. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Xiaomin Kang
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Wei Yang
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Dongxu Feng
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China.,Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Xinxin Jin
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Zhengmin Ma
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Zhuang Qian
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Tianping Xie
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Huixia Li
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Jiali Liu
- Department of Clinical Laboratory, Second Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Ruiqi Wang
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| | - Fang Li
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Danhui Li
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Hongzhi Sun
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Shufang Wu
- Center for Translational Medicine, First Affiliated Hospital of Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
65
|
Hasegawa A, Yonezawa T, Taniguchi N, Otabe K, Akasaki Y, Matsukawa T, Saito M, Neo M, Marmorstein LY, Lotz MK. Role of Fibulin 3 in Aging-Related Joint Changes and Osteoarthritis Pathogenesis in Human and Mouse Knee Cartilage. Arthritis Rheumatol 2017; 69:576-585. [PMID: 27780308 DOI: 10.1002/art.39963] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/13/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The EFEMP1 gene encoding fibulin 3 is specifically expressed in the superficial zone (SZ) of articular cartilage. The aims of this study were to examine the expression patterns of fibulin 3 in the knee joints during aging and during osteoarthritis (OA) and to determine the role of fibulin 3 in the pathogenesis of OA. METHODS Immunohistochemical analysis was performed on normal and OA knee cartilage samples from humans and mice. Experimental OA was induced in wild-type and fibulin 3-/- mice, and the severity of OA was evaluated by histologic scoring. To examine fibulin 3 function, human chondrocyte monolayer cultures were transfected with small interfering RNA (siRNA), followed by quantitative polymerase chain reaction and Western blot analyses. Human bone marrow-derived mesenchymal stem cells (BM-MSCs) were transduced with an EFEMP1 lentivirus and analyzed for markers of chondrogenesis. RESULTS Fibulin 3 was specifically expressed in the SZ of normal knee joint cartilage from humans and mice, and the expression levels declined with aging. Both aging-related OA and experimental OA were significantly more severe in fibulin 3-/- mice compared with wild-type mice. Fibulin 3 expression was high in undifferentiated human BM-MSCs and decreased during chondrogenesis. Suppression of fibulin 3 by siRNA significantly increased the expression of SOX9, type II collagen, and aggrecan in human articular chondrocytes, while overexpression of fibulin 3 inhibited chondrogenesis in BM-MSCs. CONCLUSION Fibulin 3 is specifically expressed in the SZ of articular cartilage and its expression is reduced in aging and OA. Fibulin 3 regulates differentiation of adult progenitor cells, and its aging-related decline is an early event in the pathogenesis of OA. Preventing aging-associated loss of fibulin 3 or restoring it to normal levels in SZ chondrocytes has the potential to delay or prevent the onset of OA.
Collapse
Affiliation(s)
- Akihiko Hasegawa
- The Scripps Research Institute, La Jolla, California, and Osaka Medical College, Osaka, Japan
| | - Tomo Yonezawa
- The Scripps Research Institute, La Jolla, California
| | | | - Koji Otabe
- The Scripps Research Institute, La Jolla, California
| | - Yukio Akasaki
- The Scripps Research Institute, La Jolla, California
| | | | | | | | | | - Martin K Lotz
- The Scripps Research Institute, La Jolla, California
| |
Collapse
|
66
|
Hirt J, Liton PB. Autophagy and mechanotransduction in outflow pathway cells. Exp Eye Res 2017; 158:146-153. [PMID: 27373974 PMCID: PMC5199638 DOI: 10.1016/j.exer.2016.06.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 12/19/2022]
Abstract
Because of elevations in IOP and other forces, cells in the trabecular meshwork (TM) are constantly subjected to mechanical strain. In order to preserve cellular function and regain homeostasis, cells must sense and adapt to these morphological changes. We and others have already shown that mechanical stress can trigger a broad range of responses in TM cells; however, very little is known about the strategies that TM cells use to respond to this stress, so they can adapt and survive. Autophagy, a lysosomal degradation pathway, has emerged as an important cellular homeostatic mechanism promoting cell survival and adaptation to a number of cytotoxic stresses. Our laboratory has reported the activation of autophagy in TM cells in response to static biaxial strain and high pressure. Moreover, our newest data also suggest the activation of chaperon-assisted selective autophagy, a recently identified tension-induced autophagy essential for mechanotransduction, in TM cells under cyclic mechanical stress. In this review manuscript we will discuss autophagy as part of an integrated response triggered in TM cells in response to strain, exerting a dual role in repair and mechanotransduction, and the potential effects of dysregulated in outflow pathway pathophysiology.
Collapse
Affiliation(s)
- Joshua Hirt
- Duke University, Department of Ophthalmology, Durham, NC, USA
| | - Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, USA.
| |
Collapse
|
67
|
Xue JF, Shi ZM, Zou J, Li XL. Inhibition of PI3K/AKT/mTOR signaling pathway promotes autophagy of articular chondrocytes and attenuates inflammatory response in rats with osteoarthritis. Biomed Pharmacother 2017; 89:1252-1261. [DOI: 10.1016/j.biopha.2017.01.130] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 01/21/2017] [Accepted: 01/21/2017] [Indexed: 01/15/2023] Open
|
68
|
Shen T, Alvarez-Garcia O, Li Y, Olmer M, Lotz MK. Suppression of Sestrins in aging and osteoarthritic cartilage: dysfunction of an important stress defense mechanism. Osteoarthritis Cartilage 2017; 25:287-296. [PMID: 27693501 PMCID: PMC5258682 DOI: 10.1016/j.joca.2016.09.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/31/2016] [Accepted: 09/23/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Aging is an important osteoarthritis (OA) risk factor and compromised stress defense responses may mediate this risk. The Sestrins (Sesn) promote cell survival under stress conditions and regulate AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) signaling. This study examined Sesn expression in normal and OA cartilage and functions of Sesn in chondrocytes. METHODS Sesn expression in human and mouse normal and OA cartilage was analyzed by quantitative polymerase chain reaction (PCR) and immunohistochemistry. Sesn function was investigated by using small interfering RNA (siRNA) mediated Sesn knockdown and overexpression with analysis of cell survival, gene expression, autophagy, and AMPK and mTOR activation. RESULTS Sesn mRNA levels were significantly reduced in human OA cartilage and immunohistochemistry of human and mouse OA cartilage also showed a corresponding reduction in protein levels. In cultured human chondrocytes Sesn1, 2 and 3 were expressed and increased by tunicamycin, an endoplasmic reticulum (ER) stress response inducer and 2-deoxyglucose (2DG), a metabolic stress inducer. Sesn1 and 2 were increased by tBHP, an oxidative stress inducer. Sesn knockdown by siRNA reduced chondrocyte viability under basal culture conditions and in the presence of 2DG. Sesn overexpression enhanced LC3-II formation and autophagic flux, and this was related to changes in mTOR but not AMPK activation. CONCLUSION These findings are the first to show that Sesn expression is suppressed in OA affected cartilage. Sesn support chondrocyte survival under stress conditions and promote autophagy activation through modulating mTOR activity. Suppression of Sesn in OA cartilage contributes to deficiency in an important cellular homeostasis mechanism.
Collapse
Affiliation(s)
- Tao Shen
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA,Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Shenyang 110004, People’s Republic of China
| | - Oscar Alvarez-Garcia
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA
| | - Yan Li
- Department of Orthopedic Surgery, Shengjing Hospital, China Medical University, No. 36, Sanhao Street, Shenyang 110004, People’s Republic of China
| | - Merissa Olmer
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA
| | - Martin K. Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA
| |
Collapse
|
69
|
Kim EK, Kwon JE, Lee SY, Lee EJ, Kim DS, Moon SJ, Lee J, Kwok SK, Park SH, Cho ML. IL-17-mediated mitochondrial dysfunction impairs apoptosis in rheumatoid arthritis synovial fibroblasts through activation of autophagy. Cell Death Dis 2017; 8:e2565. [PMID: 28102843 PMCID: PMC5386390 DOI: 10.1038/cddis.2016.490] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/08/2016] [Accepted: 12/06/2016] [Indexed: 02/06/2023]
Abstract
Fibroblast-like synoviocytes (FLSs) are a major cell population of the pannus that invades cartilage and bone in rheumatoid arthritis (RA). FLS resistance to apoptosis is a major characteristic of RA. The aims of this study were to investigate the effects of interleukin-17 (IL-17) and IL-17-producing T helper (Th17) cells on resistance to apoptosis in FLSs from RA patients (RA FLSs) and their roles in mitochondrial dysfunction and autophagy. Mitochondrial function was assessed in RA FLSs and FLSs from osteoarthritis patients (OA FLSs). FLSs were treated with IL-17 and their morphological features, respiratory level and mitochondrial gene expression were measured. The effects of IL-17 and Th17 cells on the relationship between autophagy and apoptosis were evaluated by measuring the expression of apoptosis-related genes using sodium nitroprusside or 3-methyladenine. The mitochondria of FLSs isolated from RA and osteoarthritis patients displayed different morphological and physiological features. RA FLSs exhibited greater autophagosome formation and greater dysfunction of mitochondrial respiration compared with OA FLSs. IL-17 induced mitochondrial dysfunction and autophagosome formation in RA FLSs, suggesting that they were resistant to apoptosis. Autophagy-related antiapoptosis induced by IL-17 was restored by inhibition of autophagy, suggesting a relationship between mitochondrial dysfunction and cell survival in RA FLSs. Th17 cells and IL-17 increased autophagy of RA FLSs by causing mitochondrial dysfunction. Our findings suggest that, in RA, interactions between RA FLSs and Th17 cells may be involved in the tumorous growth of FLSs and the formation of pannus in joints.
Collapse
Affiliation(s)
- Eun Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seocho-gu, Seoul, South Korea
| | - Jeong-Eun Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seocho-gu, Seoul, South Korea
| | - Seon-Young Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seocho-gu, Seoul, South Korea
| | - Eun-Jung Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seocho-gu, Seoul, South Korea
| | - Da Som Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seocho-gu, Seoul, South Korea
| | - Su-Jin Moon
- Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jennifer Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea.,Laboratory of Immune Network, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seocho-gu, Seoul, South Korea
| |
Collapse
|
70
|
Insights on Molecular Mechanisms of Chondrocytes Death in Osteoarthritis. Int J Mol Sci 2016; 17:ijms17122146. [PMID: 27999417 PMCID: PMC5187946 DOI: 10.3390/ijms17122146] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/05/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a joint pathology characterized by progressive cartilage degradation. Medical care is mainly based on alleviating pain symptoms. Compelling studies report the presence of empty lacunae and hypocellularity in cartilage with aging and OA progression, suggesting that chondrocyte cell death occurs and participates to OA development. However, the relative contribution of apoptosis per se in OA pathogenesis appears complex to evaluate. Indeed, depending on technical approaches, OA stages, cartilage layers, animal models, as well as in vivo or in vitro experiments, the percentage of apoptosis and cell death types can vary. Apoptosis, chondroptosis, necrosis, and autophagic cell death are described in this review. The question of cell death causality in OA progression is also addressed, as well as the molecular pathways leading to cell death in response to the following inducers: Fas, Interleukin-1β (IL-1β), Tumor Necrosis factor-α (TNF-α), leptin, nitric oxide (NO) donors, and mechanical stresses. Furthermore, the protective role of autophagy in chondrocytes is highlighted, as well as its decline during OA progression, enhancing chondrocyte cell death; the transition being mainly controlled by HIF-1α/HIF-2α imbalance. Finally, we have considered whether interfering in chondrocyte apoptosis or promoting autophagy could constitute therapeutic strategies to impede OA progression.
Collapse
|
71
|
June RK, Liu-Bryan R, Long F, Griffin TM. Emerging role of metabolic signaling in synovial joint remodeling and osteoarthritis. J Orthop Res 2016; 34:2048-2058. [PMID: 27605370 PMCID: PMC5365077 DOI: 10.1002/jor.23420] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/31/2016] [Indexed: 02/04/2023]
Abstract
Obesity and associated metabolic diseases collectively referred to as the metabolic syndrome increase the risk of skeletal and synovial joint diseases, including osteoarthritis (OA). The relationship between obesity and musculoskeletal diseases is complex, involving biomechanical, dietary, genetic, inflammatory, and metabolic factors. Recent findings illustrate how changes in cellular metabolism and metabolic signaling pathways alter skeletal development, remodeling, and homeostasis, especially in response to biomechanical and inflammatory stressors. Consequently, a better understanding of the energy metabolism of diarthrodial joint cells and tissues, including bone, cartilage, and synovium, may lead to new strategies to treat or prevent synovial joint diseases such as OA. This rationale was the basis of a workshop presented at the 2016 Annual ORS Meeting in Orlando, FL on the emerging role of metabolic signaling in synovial joint remodeling and OA. The topics we covered included (i) the relationship between metabolic syndrome and OA in clinical and pre-clinical studies; (ii) the effect of biomechanical loading on chondrocyte metabolism; (iii) the effect of Wnt signaling on osteoblast carbohydrate and amino acid metabolism with respect to bone anabolism; and (iv) the role of AMP-activated protein kinase in chondrocyte energetic and biomechanical stress responses in the context of cartilage injury, aging, and OA. Although challenges exist for measuring in vivo changes in synovial joint tissue metabolism, the findings presented herein provide multiple lines of evidence to support a central role for disrupted cellular energy metabolism in the pathogenesis of OA. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:2048-2058, 2016.
Collapse
Affiliation(s)
- Ronald K. June
- Depts. of Mechanical & Industrial Engineering and Cell Biology & Neuroscience, Montana State University, Bozeman, MT, USA
| | - Ru Liu-Bryan
- VA San Diego Healthcare System, Dept. of Medicine, University of California San Diego, San Diego, California, USA
| | - Fanxing Long
- Dept. of Orthopaedic Surgery, Dept. of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Timothy M. Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Depts. of Biochemistry and Molecular Biology, Physiology, and Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
72
|
van Loosdregt J, Rossetti M, Spreafico R, Moshref M, Olmer M, Williams GW, Kumar P, Copeland D, Pischel K, Lotz M, Albani S. Increased autophagy in CD4 + T cells of rheumatoid arthritis patients results in T-cell hyperactivation and apoptosis resistance. Eur J Immunol 2016; 46:2862-2870. [PMID: 27624289 DOI: 10.1002/eji.201646375] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 07/26/2016] [Accepted: 09/09/2016] [Indexed: 11/08/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease hallmarked by aberrant cellular homeostasis, resulting in hyperactive CD4+ T cells that are more resistant to apoptosis. Both hyperactivation and resistance to apoptosis may contribute to the pathogenicity of CD4+ T cells in the autoimmune process. A better knowledge of the mechanisms determining such impaired homeostasis could contribute significantly to both the understanding and the treatment of the disease. Here we investigated whether autophagy, is dysregulated in CD4+ T cells of RA patients, resulting in disturbed T-cell homeostasis. We demonstrate that the rate of autophagy is significantly increased in CD4+ T cells from RA patients, and that increased autophagy is also a feature of in vitro activated CD4+ T cells. The increased apoptosis resistance observed in CD4+ T cells from RA patients was significantly reversed upon autophagy inhibition. These mechanisms may contribute to RA pathogenesis, as autophagy inhibition reduced both arthritis incidence and disease severity in a mouse collagen induced arthritis mouse model. Conversely, in Atg5flox/flox -CD4-Cre+ mice, in which all T cells are autophagy deficient, T cells showed impaired activation and proliferation. These data provide novel insight into the pathogenesis of RA and underscore the relevance of autophagy as a promising therapeutic target.
Collapse
Affiliation(s)
- Jorg van Loosdregt
- Translational Research Laboratory, Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, USA.,Eureka Institute for Translational Medicine, Siracusa, Italy.,Division of Pediatrics, University Medical Center, Utrecht, The Netherlands.,Center for Molecular Medicine, University Medical Center, Utrecht, The Netherlands
| | - Maura Rossetti
- Translational Research Laboratory, Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, USA.,SingHealth Translational Immunology and Inflammation Centre, Duke-NUS Graduate Medical School, Singapore, Singapore.,Eureka Institute for Translational Medicine, Siracusa, Italy
| | - Roberto Spreafico
- Translational Research Laboratory, Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, USA.,SingHealth Translational Immunology and Inflammation Centre, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Maryam Moshref
- Translational Research Laboratory, Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, USA
| | - Merissa Olmer
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, USA
| | | | - Pavanish Kumar
- SingHealth Translational Immunology and Inflammation Centre, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Dana Copeland
- Division of Rheumatology, Scripps Clinic, San Diego, USA
| | - Ken Pischel
- Division of Rheumatology, Scripps Clinic, San Diego, USA
| | - Martin Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, USA
| | - Salvatore Albani
- Translational Research Laboratory, Inflammatory and Infectious Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, USA.,SingHealth Translational Immunology and Inflammation Centre, Duke-NUS Graduate Medical School, Singapore, Singapore.,Eureka Institute for Translational Medicine, Siracusa, Italy
| |
Collapse
|
73
|
Coughlin TR, Kennedy OD. The role of subchondral bone damage in post-traumatic osteoarthritis. Ann N Y Acad Sci 2016; 1383:58-66. [DOI: 10.1111/nyas.13261] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/19/2016] [Accepted: 08/23/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Thomas R. Coughlin
- Department of Orthopedic Surgery; New York University School of Medicine; New York New York
| | - Oran D. Kennedy
- Department of Orthopedic Surgery; New York University School of Medicine; New York New York
- Department of Mechanical and Aerospace Engineering; New York University Tandon School of Engineering; New York New York
| |
Collapse
|
74
|
Autophagy transduces physical constraints into biological responses. Int J Biochem Cell Biol 2016; 79:419-426. [PMID: 27566364 DOI: 10.1016/j.biocel.2016.08.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 12/15/2022]
Abstract
Autophagy is a fundamental cell biological process that controls the quality and quantity of the eukaryotic cytoplasm. Dysfunctional autophagy, when defective or excessive, has been linked to human pathologies ranging from neurodegenerative and infectious diseases to cancer and inflammatory diseases. Autophagy takes place at basal levels in all eukaryotic cells. The process is stimulated during metabolic, genotoxic, infectious, and hypoxic stress conditions and acts an adaptive mechanism essential for cell survival. Recent data demonstrate that changes in the mechanical cellular environment influence cell fate through the modulation of the autophagic pathway. Mechanical stimuli, such as applied forces, combine with biochemical signals to control development and physiological functions of different organs and can also contribute to the progression of various human diseases. Here we review recent findings regarding the regulation of autophagy upon three types of mechanical stress, compression, shear stress, and stretching, and discuss the potential implications of mechanical stress-induced autophagy in physiology and physiopathology.
Collapse
|
75
|
Rockel JS, Kapoor M. Autophagy: controlling cell fate in rheumatic diseases. Nat Rev Rheumatol 2016; 12:517-31. [DOI: 10.1038/nrrheum.2016.92] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
76
|
Oxidative stress, autophagy, epigenetic changes and regulation by miRNAs as potential therapeutic targets in osteoarthritis. Biochem Pharmacol 2016; 108:1-10. [DOI: 10.1016/j.bcp.2015.12.012] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/14/2015] [Indexed: 02/07/2023]
|
77
|
Ribeiro M, López de Figueroa P, Blanco FJ, Mendes AF, Caramés B. Insulin decreases autophagy and leads to cartilage degradation. Osteoarthritis Cartilage 2016; 24:731-9. [PMID: 26549531 DOI: 10.1016/j.joca.2015.10.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/16/2015] [Accepted: 10/28/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Autophagy, a key homeostasis mechanism, is defective in Osteoarthritis (OA) and Type 2 Diabetes (T2D). T2D has been proposed as a risk factor for OA. We hypothesized that diabetes impairs articular cartilage integrity by decreasing autophagy. Our objective was to investigate the effects of high glucose and insulin, characteristics of T2D, on cartilage homeostasis. METHODS Immortalized human chondrocytes (TC28a2) and primary human chondrocytes (HC) were cultured in 25 mM or 0 mM glucose and treated with insulin (10, 100, 500 nM) for 2, 6 or 24 h. Activity of LC3-II, Akt and rpS6 was evaluated by Western blotting (WB). Human cartilage explants were cultivated with 25 mM glucose and insulin (100,1000 nM) for 24 h to evaluate histopathology. MMP-13 and IL-1β expression was determined by immunohistochemistry and WB. Effects of Rapamycin (10 μM) were analyzed by WB. LC3 and rpS6 expression was determined by WB in chondrocytes from Healthy, Non Diabetic-OA and Diabetic-OA patients. RESULTS Insulin downregulates autophagy by reducing LC3 II expression and increasing Akt and rpS6 phosphorylation. Loss of proteoglycans and increased MMP-13 and IL-1β expression was observed after insulin treatment. Autophagy activation by rapamycin reversed insulin effects. Importantly, chondrocytes from diabetic-OA patients showed decreased LC3 and increased p-rpS6 expression compared to Healthy and Non-Diabetic OA patients. CONCLUSIONS These results suggest that decreased autophagy might be a mechanism by which diabetes influences cartilage degradation. Pharmacological activation of autophagy may be an effective therapeutic approach to prevent T2D-induced cartilage damage.
Collapse
Affiliation(s)
- M Ribeiro
- Grupo de Biología del Cartílago, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain; Centre for Neuroscience and Cell Biology and Faculty of Pharmacy, University of Coimbra, Portugal
| | - P López de Figueroa
- Grupo de Biología del Cartílago, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - F J Blanco
- Grupo de Biología del Cartílago, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain
| | - A F Mendes
- Centre for Neuroscience and Cell Biology and Faculty of Pharmacy, University of Coimbra, Portugal
| | - B Caramés
- Grupo de Biología del Cartílago, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Spain.
| |
Collapse
|
78
|
Li YS, Zhang FJ, Zeng C, Luo W, Xiao WF, Gao SG, Lei GH. Autophagy in osteoarthritis. Joint Bone Spine 2016; 83:143-8. [DOI: 10.1016/j.jbspin.2015.06.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/21/2015] [Indexed: 01/15/2023]
|
79
|
Shi J, Zhang C, Yi Z, Lan C. Explore the variation of MMP3, JNK, p38 MAPKs, and autophagy at the early stage of osteoarthritis. IUBMB Life 2016; 68:293-302. [PMID: 26873249 DOI: 10.1002/iub.1482] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 01/17/2016] [Indexed: 12/20/2022]
Abstract
Osteoarthritis is a chronic disease characterized by cartilage degeneration and chondrocyte apoptosis. Mitogen-activated protein kinase (MAPK) signaling pathway plays a key role in regulating OA process. Autophagy has an important effect on the OA process, and it is believed to be regulated by MAPKs. To reveal the mechanism and the effect of JNK and p38 MAPKs on matrix metalloproteinase 3 (MMP3) and autophagy in OA, the study established OA model in rabbits, used the measurement of the Osteoarthritis Research Society International scoring system to evaluate OA model, and conducted general observation, histological observation, and Western blotting of JNK, phosphorylate-JNK (P-JNK), p38, phosphorylate-p38 (P-p38), MMP3, and light-chain 3 (LC3)-II/LC3-I to explore the variation of JNK, p38 MAPKs, and autophagy at the early stage of OA. With OA progressing at the early stage, MMP3, P-p38, and P-JNK were gradually upregulated from the baseline to the peak in study groups when compared with the control group; JNK and p38 variated of turbulence without statistical difference; and LC3-II/LC3-I had a decreasing tendency from the 0- to 15-day group. This study identifies that compromised autophagy may be related to the OA progress and that JNK and p38 MAPKs have positive regulation on MMP3 and negative regulation on autophagy. It also implicates a new therapeutic strategy for OA and other degenerate diseases based on selective MAPK inhibitors, reduction of MMP3, and autophagy.
Collapse
Affiliation(s)
- Jie Shi
- Department of Rehabilitation Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Changjie Zhang
- Department of Rehabilitation Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Zhongjie Yi
- Department of General Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Chunna Lan
- Department of Rehabilitation Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
80
|
Monosodium Urate Crystal-Induced Chondrocyte Death via Autophagic Process. Int J Mol Sci 2015; 16:29265-77. [PMID: 26670233 PMCID: PMC4691108 DOI: 10.3390/ijms161226164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 12/19/2022] Open
Abstract
Monosodium urate (MSU) crystals, which are highly precipitated in the joint cartilage, increase the production of cartilage-degrading enzymes and pro-inflammatory mediators in cartilage, thereby leading to gouty inflammation and joint damage. In this study, we investigated the effect of MSU crystals on the viability of human articular chondrocytes and the mechanism of MSU crystal-induced chondrocyte death. MSU crystals significantly decreased the viability of primary chondrocytes in a time- and dose-dependent manner. DNA fragmentation was observed in a culture medium of MSU crystal-treated chondrocytes, but not in cell lysates. MSU crystals did not activate caspase-3, a marker of apoptosis, compared with actinomycin D and TNF-α-treated cells. MSU crystals did not directly affect the expression of endoplasmic reticulum (ER) stress markers at the mRNA and protein levels. However, MSU crystals significantly increased the LC3-II level in a time-dependent manner, indicating autophagy activation. Moreover, MSU crystal-induced autophagy and subsequent chondrocyte death were significantly inhibited by 3-methyladenine, a blocker of autophagosomes formation. MSU crystals activated autophagy via inhibition of phosporylation of the Akt/mTOR signaling pathway. These results demonstrate that MSU crystals may cause the death of chondrocytes through the activation of the autophagic process rather than apoptosis or ER stress.
Collapse
|
81
|
Wang X, Qi H, Wang Q, Zhu Y, Wang X, Jin M, Tan Q, Huang Q, Xu W, Li X, Kuang L, Tang Y, Du X, Chen D, Chen L. FGFR3/fibroblast growth factor receptor 3 inhibits autophagy through decreasing the ATG12-ATG5 conjugate, leading to the delay of cartilage development in achondroplasia. Autophagy 2015; 11:1998-2013. [PMID: 26491898 PMCID: PMC4824585 DOI: 10.1080/15548627.2015.1091551] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 08/26/2015] [Accepted: 09/03/2015] [Indexed: 01/15/2023] Open
Abstract
FGFR3 (fibroblast growth factor receptor 3) is a negative regulator of endochondral ossification. Gain-of-function mutations in FGFR3 are responsible for achondroplasia, the most common genetic form of dwarfism in humans. Autophagy, an evolutionarily conserved catabolic process, maintains chondrocyte viability in the growth plate under stress conditions, such as hypoxia and nutritional deficiencies. However, the role of autophagy and its underlying molecular mechanisms in achondroplasia remain elusive. In this study, we found activated FGFR3 signaling inhibited autophagic activity in chondrocytes, both in vivo and in vitro. By employing an embryonic bone culture system, we demonstrated that treatment with autophagy inhibitor 3-MA or chloroquine led to cartilage growth retardation, which mimics the effect of activated-FGFR3 signaling on chondrogenesis. Furthermore, we found that FGFR3 interacted with ATG12-ATG5 conjugate by binding to ATG5. More intriguingly, FGFR3 signaling was found to decrease the protein level of ATG12-ATG5 conjugate. Consistently, using in vitro chondrogenic differentiation assay system, we showed that the ATG12-ATG5 conjugate was essential for the viability and differentiation of chondrocytes. Transient transfection of ATG5 partially rescued FGFR3-mediated inhibition on chondrocyte viability and differentiation. Our findings reveal that FGFR3 inhibits the autophagic activity by decreasing the ATG12-ATG5 conjugate level, which may play an essential role in the pathogenesis of achondroplasia.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Huabing Qi
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Quan Wang
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Ying Zhu
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Xianxing Wang
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Qiaoyan Tan
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Qizhao Huang
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Wei Xu
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Xiaogang Li
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Liang Kuang
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Yubing Tang
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| | - Di Chen
- Department of Biochemistry; Rush University Medical Center; Chicago, IL USA
| | - Lin Chen
- Center of Bone Metabolism and Repair (CBMR); Trauma Center; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing,China
- State Key Laboratory of Trauma; Burns and Combined Injury; Third Military Medical University; Chongqing, China
- Department of Rehabilitation Medicine; Institute of Surgery Research; Daping Hospital; Third Military Medical University; Chongqing, China
| |
Collapse
|
82
|
Inflammation and intracellular metabolism: new targets in OA. Osteoarthritis Cartilage 2015; 23:1835-42. [PMID: 26521729 PMCID: PMC4668929 DOI: 10.1016/j.joca.2014.12.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/17/2014] [Indexed: 02/02/2023]
Abstract
Articular cartilage degeneration is hallmark of osteoarthritis (OA). Low-grade chronic inflammation in the joint can promote OA progression. Emerging evidence indicates that bioenergy sensors couple metabolism with inflammation to switch physiological and clinical phenotypes. Changes in cellular bioenergy metabolism can reprogram inflammatory responses, and inflammation can disturb cellular energy balance and increase cell stress. AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1) are two critical bioenergy sensors that regulate energy balance at both cellular and whole-body levels. Dysregulation of AMPK and SIRT1 has been implicated in diverse human diseases and aging. This review reveals recent findings on the role of AMPK and SIRT1 in joint tissue homeostasis and OA, with a focus on how AMPK and SIRT1 in articular chondrocytes modulate intracellular energy metabolism during stress responses (e.g., inflammatory responses) and how these changes dictate specific effector functions, and discusses translational significance of AMPK and SIRT1 as new therapeutic targets for OA.
Collapse
|
83
|
Chondrocyte Apoptosis in the Pathogenesis of Osteoarthritis. Int J Mol Sci 2015; 16:26035-54. [PMID: 26528972 PMCID: PMC4661802 DOI: 10.3390/ijms161125943] [Citation(s) in RCA: 586] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/03/2015] [Accepted: 10/21/2015] [Indexed: 11/16/2022] Open
Abstract
Apoptosis is a highly-regulated, active process of cell death involved in development, homeostasis and aging. Dysregulation of apoptosis leads to pathological states, such as cancer, developmental anomalies and degenerative diseases. Osteoarthritis (OA), the most common chronic joint disease in the elderly population, is characterized by progressive destruction of articular cartilage, resulting in significant disability. Because articular cartilage depends solely on its resident cells, the chondrocytes, for the maintenance of extracellular matrix, the compromising of chondrocyte function and survival would lead to the failure of the articular cartilage. The role of subchondral bone in the maintenance of proper cartilage matrix has been suggested as well, and it has been proposed that both articular cartilage and subchondral bone interact with each other in the maintenance of articular integrity and physiology. Some investigators include both articular cartilage and subchondral bone as targets for repairing joint degeneration. In late-stage OA, the cartilage becomes hypocellular, often accompanied by lacunar emptying, which has been considered as evidence that chondrocyte death is a central feature in OA progression. Apoptosis clearly occurs in osteoarthritic cartilage; however, the relative contribution of chondrocyte apoptosis in the pathogenesis of OA is difficult to evaluate, and contradictory reports exist on the rate of apoptotic chondrocytes in osteoarthritic cartilage. It is not clear whether chondrocyte apoptosis is the inducer of cartilage degeneration or a byproduct of cartilage destruction. Chondrocyte death and matrix loss may form a vicious cycle, with the progression of one aggravating the other, and the literature reveals that there is a definite correlation between the degree of cartilage damage and chondrocyte apoptosis. Because current treatments for OA act only on symptoms and do not prevent or cure OA, chondrocyte apoptosis would be a valid target to modulate cartilage degeneration.
Collapse
|
84
|
Bouderlique T, Vuppalapati KK, Newton PT, Li L, Barenius B, Chagin AS. Targeted deletion of Atg5 in chondrocytes promotes age-related osteoarthritis. Ann Rheum Dis 2015; 75:627-31. [PMID: 26438374 PMCID: PMC4789686 DOI: 10.1136/annrheumdis-2015-207742] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 08/11/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVES It has been suggested that the lysosomal recycling process called macro-autophagy plays a role in osteoarthritis development. We thus decided to genetically ablate the autophagy-indispensable Atg5 gene specifically in chondrocytes and analyse the development of osteoarthritis upon aging and in a post-traumatic model. METHODS Mice lacking the Atg5 gene in their chondrocytes (Atg5cKO) were generated by crossing Atg5-floxed mice with transgenic mice that expressed cre recombinase driven by the collagen type 2 promoter. Animals were analysed at the age of 2, 6 and 12 months for age-related osteoarthritis or underwent mini-open partial medial meniscectomy at 2 months of age and were analysed 1 or 2 months after surgery. We evaluated osteoarthritis using the Osteoarthritis Research Society International (OARSI) scoring on safranin-O-stained samples. Cell death was evaluated by terminal deoxy-nucleotidyl-transferase-mediated deoxy-UTP nick end labelling (TUNEL) and by immunostaining of cleaved caspases. RESULTS We observed the development of osteoarthritis in Atg5cKO mice with aging including fibrillation and loss of proteoglycans, which was particularly severe in males. The ablation of Atg5 was associated with an increased cell death as assessed by TUNEL, cleaved caspase 3 and cleaved caspase 9. Surprisingly, no difference in the development of post-traumatic osteoarthritis was observed between Atg5cKO and control mice. CONCLUSIONS Autophagy protects from age-related osteoarthritis by facilitating chondrocyte survival.
Collapse
Affiliation(s)
- Thibault Bouderlique
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Karuna K Vuppalapati
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Phillip T Newton
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Lei Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Björn Barenius
- Orthopaedic Section, Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
85
|
Non-invasive mouse models of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2015; 23:1627-38. [PMID: 26003950 PMCID: PMC4577460 DOI: 10.1016/j.joca.2015.05.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 04/20/2015] [Accepted: 05/10/2015] [Indexed: 02/02/2023]
Abstract
Animal models of osteoarthritis (OA) are essential tools for investigating the development of the disease on a more rapid timeline than human OA. Mice are particularly useful due to the plethora of genetically modified or inbred mouse strains available. The majority of available mouse models of OA use a joint injury or other acute insult to initiate joint degeneration, representing post-traumatic osteoarthritis (PTOA). However, no consensus exists on which injury methods are most translatable to human OA. Currently, surgical injury methods are most commonly used for studies of OA in mice; however, these methods may have confounding effects due to the surgical/invasive injury procedure itself, rather than the targeted joint injury. Non-invasive injury methods avoid this complication by mechanically inducing a joint injury externally, without breaking the skin or disrupting the joint. In this regard, non-invasive injury models may be crucial for investigating early adaptive processes initiated at the time of injury, and may be more representative of human OA in which injury is induced mechanically. A small number of non-invasive mouse models of PTOA have been described within the last few years, including intra-articular fracture of tibial subchondral bone, cyclic tibial compression loading of articular cartilage, and anterior cruciate ligament (ACL) rupture via tibial compression overload. This review describes the methods used to induce joint injury in each of these non-invasive models, and presents the findings of studies utilizing these models. Altogether, these non-invasive mouse models represent a unique and important spectrum of animal models for studying different aspects of PTOA.
Collapse
|
86
|
Caramés B, Olmer M, Kiosses WB, Lotz MK. The relationship of autophagy defects to cartilage damage during joint aging in a mouse model. Arthritis Rheumatol 2015; 67:1568-76. [PMID: 25708836 DOI: 10.1002/art.39073] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 02/10/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Aging is a main risk factor for osteo arthritis (OA), the most prevalent musculoskeletal disorder. Defects in autophagy, an essential cellular homeostasis mechanism, have recently been observed in OA articular cartilage. The objectives of this study were to establish the constitutive level of autophagy activation in normal cartilage and to monitor the temporal relationship between changes in autophagy and aging-related degradation of cartilage in a mouse model. METHODS In GFP-LC3-transgenic mice, green fluorescent protein (GFP)-light chain 3 (LC3) is ubiquitously expressed, and the accumulation of GFP puncta, representing autophagosomes, was quantified by confocal microscopy as a measure of autophagy activation. Expression of the autophagy proteins autophagy-related protein 5 (ATG-5) and microtubule-associated protein 1 light chain 3 (LC3) was analyzed by immunohistochemistry. Cartilage cellularity, apoptotic cell death, and cartilage structural damage and changes in synovium and bone were examined by histology and immunohistochemistry. RESULTS Basal autophagy activation was detected in liver and knee articular cartilage from young (6-month-old) mice, with higher levels in cartilage than in liver in the same animals. In 28-month-old mice, there was a statistically significant reduction in the total number of autophagic vesicles per cell (P < 0.01) and in the total area of vesicles per cell (P < 0.01) in the articular cartilage as compared to that from young 6-month-old mice. With increasing age, the expression of ATG-5 and LC3 decreased, and this was followed by a reduction in cartilage cellularity and an increase in the apoptosis marker poly(ADP-ribose) polymerase p85. Cartilage structural damage progressed in an age-dependent manner subsequent to the autophagy changes. CONCLUSION Autophagy is constitutively activated in normal cartilage. This is compromised with aging and precedes cartilage cell death and structural damage.
Collapse
Affiliation(s)
- Beatriz Caramés
- The Scripps Research Institute La Jolla California and Instituto de Investigación Biomédica de A Coruña Complexo Hospitalario Universitario de A Coruña SERGAS and Universidade da Coruña, A Coruña, Spain
| | - Merissa Olmer
- The Scripps Research Institute, La Jolla, California
| | | | - Martin K Lotz
- The Scripps Research Institute, La Jolla, California
| |
Collapse
|
87
|
Liton PB. The autophagic lysosomal system in outflow pathway physiology and pathophysiology. Exp Eye Res 2015; 144:29-37. [PMID: 26226231 DOI: 10.1016/j.exer.2015.07.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/06/2015] [Accepted: 07/22/2015] [Indexed: 12/19/2022]
Abstract
Malfunction of the trabecular meshwork (TM)/schlemm's canal (SC) conventional outflow pathway is associated with elevated intraocular pressure (IOP) and, therefore, increased risk of developing glaucoma, a potentially blinding disease affecting more than 70 million people worldwide. This TM/SC tissue is subjected to different types of stress, including mechanical, oxidative, and phagocytic stress. Long-term exposure to these stresses is believed to lead to a progressive accumulation of damaged cellular and tissue structures causing permanent alterations in the tissue physiology, and contribute to the pathologic increase in aqueous humor (AH) outflow resistance. Autophagy is emerging as an essential cellular survival mechanism against a variety of stressors. In addition to performing basal functions, autophagy acts as a cellular survival pathway and represents an essential mechanism by which organisms can adapt to acute stress conditions and repair stress-induced damage. A decline in autophagy has been observed in most tissues with aging and has been considered responsible, at least in part, for the accumulation of damaged cellular components in almost all tissues of aging organisms. Dysfunction in the autophagy pathway is associated with several human diseases, from infectious diseases to cancer and neurodegeneration. In this review, we will summarize our current knowledge of the emerging roles of autophagy in outflow tissue physiology and pathophysiology, including novel evidence suggesting compromised autophagy in the glaucomatous outflow pathway.
Collapse
Affiliation(s)
- Paloma B Liton
- Duke University, Department of Ophthalmology, Durham, NC, USA.
| |
Collapse
|
88
|
Jiang LB, Cao L, Dong J. Does autophagy activation really inhibit calcification of endplate chondrocytes under intermittent cyclic mechanical tension? Bone 2015; 75:240-1. [PMID: 25445444 DOI: 10.1016/j.bone.2014.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/26/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Li-Bo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu Cao
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Jian Dong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
89
|
López de Figueroa P, Lotz MK, Blanco FJ, Caramés B. Autophagy activation and protection from mitochondrial dysfunction in human chondrocytes. Arthritis Rheumatol 2015; 67:966-76. [PMID: 25605458 DOI: 10.1002/art.39025] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 01/06/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Autophagy is a key pathway of cellular homeostasis for removing damaged macromolecules and organelles, including mitochondria. Recent studies indicate that activation of autophagy is defective in aging and osteoarthritis (OA), contributing to cell death and tissue damage. In addition, there is increasing evidence that mitochondrial dysfunction plays an important role in OA pathogenesis. The objective of this study was to determine whether activation of autophagy protects against mitochondrial dysfunction in human chondrocytes. METHODS Human chondrocytes were treated with oligomycin, an inhibitor of mitochondrial respiratory chain complex V. Autophagy activation was analyzed by determination of light chain 3 membrane-bound form II (LC3-II), a marker of autophagosome formation. To investigate whether autophagy protects from mitochondrial dysfunction, autophagy was induced by rapamycin, the selective inhibitor of mammalian target of rapamycin complex 1 (mTORC-1), and by torin 1, the inhibitor of mTORC-1 and mTORC-2. Small interfering autophagy-related 5 was used to evaluate the role of autophagy in mitochondrial dysfunction. RESULTS Mitochondrial dysfunction was induced by treatment with oligomycin, which significantly decreased mitochondrial membrane potential (ΔΨm). This was associated with increased production of reactive oxygen species and cell death. Autophagy activation, as reflected by LC3-II, was decreased in a time-dependent manner. To evaluate whether autophagy regulates mitochondrial function, chondrocytes were pretreated with rapamycin and torin 1 before oligomycin. Autophagy activation significantly protected against mitochondrial dysfunction. Conversely, genetic inhibition of autophagy induced significant mitochondrial function defects. CONCLUSION Our data highlight the role of autophagy as a critical protective mechanism against mitochondrial dysfunction. Pharmacologic interventions that enhance autophagy may have chondroprotective activity in cartilage degenerative processes such as OA.
Collapse
Affiliation(s)
- Paloma López de Figueroa
- Instituto de Investigación Biomédica de A Coruña, Complexo Hospitalario Universitario de A Coruña, SERGAS, and Universidade da Coruña, A Coruña, Spain
| | | | | | | |
Collapse
|
90
|
Emerging targets in osteoarthritis therapy. Curr Opin Pharmacol 2015; 22:51-63. [PMID: 25863583 DOI: 10.1016/j.coph.2015.03.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/17/2015] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a destructive joint disease in which the initiation may be attributed to direct injury and mechanical disruption of joint tissues, but the progressive changes are dependent on active cell-mediated processes that can be observed or inferred during the generally long time-course of the disease. Based on clinical observations and experimental studies, it is now recognized a that it is possible for individual patients to exhibit common sets of symptoms and structural abnormalities due to distinct pathophysiological pathways that act independently or in combination. Recent research that has focused on the underlying mechanisms involving biochemical cross talk among the cartilage, synovium, bone, and other joint tissues within a background of poorly characterized genetic factors will be addressed in this review.
Collapse
|
91
|
Vasheghani F, Zhang Y, Li YH, Blati M, Fahmi H, Lussier B, Roughley P, Lagares D, Endisha H, Saffar B, Lajeunesse D, Marshall WK, Rampersaud YR, Mahomed NN, Gandhi R, Pelletier JP, Martel-Pelletier J, Kapoor M. PPARγ deficiency results in severe, accelerated osteoarthritis associated with aberrant mTOR signalling in the articular cartilage. Ann Rheum Dis 2015; 74:569-78. [PMID: 25573665 PMCID: PMC4345902 DOI: 10.1136/annrheumdis-2014-205743] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 11/18/2014] [Accepted: 12/07/2014] [Indexed: 11/15/2022]
Abstract
OBJECTIVES We have previously shown that peroxisome proliferator-activated receptor gamma (PPARγ), a transcription factor, is essential for the normal growth and development of cartilage. In the present study, we created inducible cartilage-specific PPARγ knockout (KO) mice and subjected these mice to the destabilisation of medial meniscus (DMM) model of osteoarthritis (OA) to elucidate the specific in vivo role of PPARγ in OA pathophysiology. We further investigated the downstream PPARγ signalling pathway responsible for maintaining cartilage homeostasis. METHODS Inducible cartilage-specific PPARγ KO mice were generated and subjected to DMM model of OA. We also created inducible cartilage-specific PPARγ/mammalian target for rapamycin (mTOR) double KO mice to dissect the PPARγ signalling pathway in OA. RESULTS Compared with control mice, PPARγ KO mice exhibit accelerated OA phenotype with increased cartilage degradation, chondrocyte apoptosis, and the overproduction of OA inflammatory/catabolic factors associated with the increased expression of mTOR and the suppression of key autophagy markers. In vitro rescue experiments using PPARγ expression vector reduced mTOR expression, increased expression of autophagy markers and reduced the expression of OA inflammatory/catabolic factors, thus reversing the phenotype of PPARγ KO mice chondrocytes. To dissect the in vivo role of mTOR pathway in PPARγ signalling, we created and subjected PPARγ-mTOR double KO mice to the OA model to see if the genetic deletion of mTOR in PPARγ KO mice (double KO) can rescue the accelerated OA phenotype observed in PPARγ KO mice. Indeed, PPARγ-mTOR double KO mice exhibit significant protection/reversal from OA phenotype. SIGNIFICANCE PPARγ maintains articular cartilage homeostasis, in part, by regulating mTOR pathway.
Collapse
Affiliation(s)
- Faezeh Vasheghani
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Yue Zhang
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network (UHN), Toronto, Ontario, Canada
| | - Ying-Hua Li
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network (UHN), Toronto, Ontario, Canada
| | - Meryem Blati
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Bertrand Lussier
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, St-Hyacinthe, Quebec, Canada
| | - Peter Roughley
- Genetics Unit, Shriners Hospital, McGill University, Montreal, Quebec, Canada
| | - David Lagares
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Massachusetts, USA
| | - Helal Endisha
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network (UHN), Toronto, Ontario, Canada
| | - Bahareh Saffar
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network (UHN), Toronto, Ontario, Canada
| | - Daniel Lajeunesse
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Wayne K Marshall
- Division of Orthopedics, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Y Raja Rampersaud
- Division of Orthopedics, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Nizar N Mahomed
- Division of Orthopedics, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Rajiv Gandhi
- Division of Orthopedics, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Unit, University of Montreal Hospital Research Centre (CRCHUM), Montreal, Quebec, Canada
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network (UHN), Toronto, Ontario, Canada
- Division of Orthopedics, Toronto Western Hospital, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
92
|
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by the progressive loss of articular cartilage, remodeling of the subchondral bone, and synovial inflammation. Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that controls critical cellular processes such as growth, proliferation, and protein synthesis. Recent studies suggest that mTOR plays a vital role in cartilage growth and development and in altering the articular cartilage homeostasis as well as contributing to the process of cartilage degeneration associated with OA. Both pharmacological inhibition and genetic deletion of mTOR have been shown to reduce the severity of OA in preclinical mouse models. In this review article, we discuss the roles of mTOR in cartilage development, in maintaining articular cartilage homeostasis, and its potential as an OA therapeutic target.
Collapse
Affiliation(s)
- Bandna Pal
- Division of Genetics and Development, The Toronto Western Research Institute, Toronto Western Hospital, The University Health Network (UHN), 60 Leonard Avenue, Toronto, ON M5T 2S8 Canada
| | - Helal Endisha
- Division of Genetics and Development, The Toronto Western Research Institute, Toronto Western Hospital, The University Health Network (UHN), 60 Leonard Avenue, Toronto, ON M5T 2S8 Canada
| | - Yue Zhang
- Division of Genetics and Development, The Toronto Western Research Institute, Toronto Western Hospital, The University Health Network (UHN), 60 Leonard Avenue, Toronto, ON M5T 2S8 Canada
| | - Mohit Kapoor
- Division of Genetics and Development, The Toronto Western Research Institute, Toronto Western Hospital, The University Health Network (UHN), 60 Leonard Avenue, Toronto, ON M5T 2S8 Canada
- Department of Surgery, University of Toronto, Toronto, ON Canada
- Division of Orthopaedics, Toronto Western Hospital, Toronto, ON Canada
| |
Collapse
|
93
|
Matsuzaki T, Matsushita T, Tabata Y, Saito T, Matsumoto T, Nagai K, Kuroda R, Kurosaka M. Intra-articular administration of gelatin hydrogels incorporating rapamycin–micelles reduces the development of experimental osteoarthritis in a murine model. Biomaterials 2014; 35:9904-9911. [DOI: 10.1016/j.biomaterials.2014.08.041] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/05/2014] [Indexed: 12/19/2022]
|
94
|
Takayama K, Kawakami Y, Kobayashi M, Greco N, Cummins JH, Matsushita T, Kuroda R, Kurosaka M, Fu FH, Huard J. Local intra-articular injection of rapamycin delays articular cartilage degeneration in a murine model of osteoarthritis. Arthritis Res Ther 2014; 16:482. [PMID: 25403236 PMCID: PMC4269094 DOI: 10.1186/s13075-014-0482-4] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2014] [Accepted: 11/05/2014] [Indexed: 02/07/2023] Open
Abstract
Introduction Recent studies have revealed that rapamycin activates autophagy in human chondrocytes preventing the development of osteoarthritis (OA) like changes in vitro, while the systemic injection of rapamycin reduces the severity of experimental osteoarthritis in a murine model of OA in vivo. Since the systemic use of rapamycin is associated with numerous side effects, the goal of the current study was to examine the beneficial effect of local intra-articular injection of rapamycin in a murine model of OA and to elucidate the mechanism of action of rapamycin on articular cartilage. Methods Destabilization of the medial meniscus (DMM) was performed on 10-week-old male mice to induce OA. Intra-articular injections of 10 μl of rapamycin (10 μM) were administered twice weekly for 8 weeks. Articular cartilage damage was analyzed by histology using a semi-quantitative scoring system at 8 and 12 weeks after surgery. Mammalian target of rapamycin (mTOR), light chain 3 (LC3), vascular endothelial growth factor (VEGF), collagen, type X alpha 1 (COL10A1), and matrix metallopeptidase 13 (MMP13) expressions were analyzed by immunohistochemistry. VEGF, COL10A1, and MMP13 expressions were further examined via quantitative RT-PCR (qPCR). Results Intra-articular injection of rapamycin significantly reduced the severity of articular cartilage degradation at 8 and 12 weeks after DMM surgery. A reduction in mTOR expression and the activation of LC3 (an autophagy marker) in the chondrocytes was observed in the rapamycin treated mice. Rapamycin treatment also reduced VEGF, COL10A1, and MMP13 expressions at 8 and 12 weeks after DMM surgery. Conclusion These results demonstrate that the intra-articular injection of rapamycin could reduce mTOR expression, leading to a delay in articular cartilage degradation in our OA murine model. Our observations suggest that local intra-articular injection of rapamycin could represent a potential therapeutic approach to prevent OA.
Collapse
|
95
|
Defective autophagy in chondrocytes with Kashin-Beck disease but higher than osteoarthritis. Osteoarthritis Cartilage 2014; 22:1936-46. [PMID: 25168363 DOI: 10.1016/j.joca.2014.08.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 08/09/2014] [Accepted: 08/18/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study was undertaken to monitor autophagy in chondrocytes with Kashin-Beck disease (KBD) and osteoarthritis (OA). METHODS The identification and quantification of autophagy were morphologically visualized by transmission electron microscopy (TEM), together with immunohistochemical localization of Beclin1 and LC3 in cartilage, and immunoblotting of cellular Beclin1, LC3 and p62/SQSTM1 in the normal, KBD and OA groups. Sequentially, regulated-autophagy genes (ATG) were analyzed by IPA software and validated by quantitative real-time polymerase chain reaction (qRT-PCR). Cytotoxicity of cell death was measured by fluorescence detection and flow cytometry (FCM). The co-localization and measurement of autophagy and mitochondria/reactive oxygen species (ROS) were carried out. RESULTS KBD chondrocytes exhibited a variety of abnormal cellular contents including nuclei, mitochondrial, glycogen deposits and microfilaments, and OA chondrocytes mainly presented swelled endocytoplasmic reticulum (ER). Beclin1 and LC3 were reduced both in KBD and OA compared with normal controls; however, the two proteins and p62 in KBD were in a higher level than OA. Simultaneously, KBD chondrocytes showed 45 genes that were different from normal controls and 92 genes different from OA, whose functions were mainly involved in cell morphology, cellular functions, cell death and survival. Autophagy was negatively correlated with apoptosis in the three kinds of chondrocytes, and the rates decreased when autophagy was induced by rapamycin. Similarly, KBD and OA chondrocytes showed lower autophagy and higher ROS production compared with the normal chondrocytes. CONCLUSION Autophagy was defective in KBD chondrocytes, but it was higher than in OA. The insufficient autophagy may be associated with apoptosis and mitochondrial change in the pathogenesis of KBD and OA.
Collapse
|
96
|
Alvarez-Garcia O, Rogers NH, Smith RG, Lotz MK. Palmitate has proapoptotic and proinflammatory effects on articular cartilage and synergizes with interleukin-1. Arthritis Rheumatol 2014; 66:1779-88. [PMID: 24591481 DOI: 10.1002/art.38399] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 02/04/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Obesity is a major risk factor for the development of osteoarthritis (OA) that is associated with a state of low-grade inflammation and increased circulating levels of adipokines and free fatty acids (FFAs). The aim of this study was to analyze the effects of saturated (palmitate) and monounsaturated (oleate) FFAs on articular chondrocytes, synoviocytes, and cartilage. METHODS Human articular chondrocytes and fibroblast-like synoviocytes obtained from young healthy donors and OA chondrocytes from patients undergoing total knee replacement surgery were treated with palmitate or oleate alone or in combination with interleukin-1β (IL-1β). Cell viability, caspase activation, and gene expression of proinflammatory factors, extracellular matrix (ECM) proteins, and proteases were studied. In addition, chondrocyte viability, IL-6 production, and matrix damage were assessed in bovine and human articular cartilage explants cultured with FFAs in the presence or absence of IL-1β. RESULTS Palmitate, but not oleate, induced caspase activation and cell death in IL-1β-stimulated normal chondrocytes, and up-regulated the expression of IL-6 and cyclooxygenase 2 in chondrocytes and fibroblast-like synoviocytes through Toll-like receptor 4 (TLR-4) signaling. In cartilage explants, palmitate induced chondrocyte death, IL-6 release, and ECM degradation. Palmitate synergized with IL-1β in stimulating proapoptotic and proinflammatory cellular responses. Pharmacologic inhibition of caspases or TLR-4 signaling reduced palmitate and IL-1β induced cartilage damage. CONCLUSION Palmitate acts as a proinflammatory and catabolic factor that, in synergy with IL-1β, induces chondrocyte apoptosis and articular cartilage breakdown. Collectively, our data suggest that elevated levels of saturated FFAs that are often found in patients who are obese may contribute to the pathogenesis of OA.
Collapse
|
97
|
Xu HG, Yu YF, Zheng Q, Zhang W, Wang CD, Zhao XY, Tong WX, Wang H, Liu P, Zhang XL. Autophagy protects end plate chondrocytes from intermittent cyclic mechanical tension induced calcification. Bone 2014; 66:232-9. [PMID: 24970040 DOI: 10.1016/j.bone.2014.06.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 06/06/2014] [Accepted: 06/17/2014] [Indexed: 01/08/2023]
Abstract
Calcification of end plate chondrocytes is a major cause of intervertebral disc (IVD) degeneration. However, the underlying molecular mechanism of end plate chondrocyte calcification is still unclear. The aim of this study was to clarify whether autophagy in end plate chondrocytes could protect the calcification of end plate chondrocytes. Previous studies showed that intermittent cyclic mechanical tension (ICMT) contributes to the calcification of end plate chondrocytes in vitro. While autophagy serves as a cell survival mechanism, the relationship of autophagy and induced end plate chondrocyte calcification by mechanical tension in vitro is unknown. Thus, we investigated autophagy, the expression of the autophagy genes, Beclin-1 and LC3, and rat end plate chondrocyte calcification by ICMT. The viability of end plate chondrocytes was examined using the LIVE/DEAD viability/cytotoxicity kit. The reverse transcription-polymerase chain reaction and western blotting were used to detect the expression of Beclin-1; LC3; type I, II and X collagen; aggrecan; and Sox-9 genes. Immunofluorescent and fluorescent microscopy showed decreased autophagy in the 10- and 20-day groups loaded with ICMT. Additionally, Alizarin red and alkaline phosphatase staining detected the palpable calcification of end plate chondrocytes after ICMT treatment. We found that increased autophagy induced by short-term ICMT treatment was accompanied by an insignificant calcification of end plate chondrocytes. To the contrary, the suppressive autophagy inhibited by long-term ICMT was accompanied by a more significant calcification. The process of calcification induced by ICMT was partially resisted by increased autophagy activity induced by rapamycin, implicating that autophagy may prevent end plate chondrocyte calcification.
Collapse
Affiliation(s)
- Hong-guang Xu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, China.
| | - Yun-fei Yu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, China
| | - Quan Zheng
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, China
| | - Wei Zhang
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, China
| | - Chuang-dong Wang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM), 200025, China
| | - Xiao-yn Zhao
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM), 200025, China
| | - Wen-xue Tong
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM), 200025, China
| | - Hong Wang
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, China
| | - Ping Liu
- Department of Orthopedic Surgery, Yijishan Hospital, Wannan Medical College, Wuhu, Anhui 241001, China
| | - Xiao-ling Zhang
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM), 200025, China.
| |
Collapse
|
98
|
Wu P, Holguin N, Silva MJ, Fu M, Liao W, Sandell LJ. Early response of mouse joint tissue to noninvasive knee injury suggests treatment targets. Arthritis Rheumatol 2014; 66:1256-65. [PMID: 24470303 DOI: 10.1002/art.38375] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 01/16/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Joint trauma can lead to a spectrum of acute lesions, including cartilage degradation, ligament or meniscus tears, and synovitis, all potentially associated with osteoarthritis (OA). This study was undertaken to generate and validate a murine model of knee joint trauma following noninvasive controlled injurious compression in vivo. METHODS The right knees of 8-week-old mice were placed in a hyperflexed position and subjected to compressive joint loading at 1 of 3 peak forces (3N, 6N, or 9N) for 60 cycles in a single loading period and harvested on days 5, 9, and 14 after loading (n = 3-5 for each time point and for each loading). The left knees were not loaded and were used as the contralateral control. Histologic, immunohistochemical, and enzyme-linked immunosorbent assay analyses were performed to evaluate acute pathologic features in chondrocyte viability, cartilage matrix metabolism, synovial reaction, and serum cartilage oligomeric matrix protein (COMP) levels. RESULTS Acute joint pathology was associated with increased injurious loads. All loading regimens induced chondrocyte apoptosis, cartilage matrix degradation, disruption of cartilage collagen fibril arrangement, and increased levels of serum COMP. We also observed that 6N loading induced mild synovitis by day 5, whereas at 9N, with tearing of the anterior cruciate ligament, severe posttraumatic synovitis and ectopic cartilage formation were observed. CONCLUSION We have established a murine model of knee joint trauma with different degrees of overloading in vivo. Our results suggest that immediate therapies particularly targeted to apoptosis and synovial cell proliferation could affect the acute posttraumatic reaction to potentially limit chronic consequences and OA.
Collapse
Affiliation(s)
- P Wu
- Washington University School of Medicine, St. Louis, Missouri; First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
99
|
Sherwood JC, Bertrand J, Eldridge SE, Dell'Accio F. Cellular and molecular mechanisms of cartilage damage and repair. Drug Discov Today 2014; 19:1172-7. [PMID: 24880104 DOI: 10.1016/j.drudis.2014.05.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/20/2014] [Indexed: 01/18/2023]
Abstract
Cartilage breakdown is the disabling outcome of rheumatic diseases, whether prevalently inflammatory such as rheumatoid arthritis or prevalently mechanical such as osteoarthritis (OA). Despite the differences between immune-mediated arthritides and OA, common mechanisms drive cartilage breakdown. Inflammation, chondrocyte phenotype and homeostatic mechanisms have recently been the focus of research and will be summarised in this review.
Collapse
Affiliation(s)
- Joanna C Sherwood
- Barts and The London, Queen Mary University of London School of Medicine and Dentistry, William Harvey Research Institute, London, UK
| | - Jessica Bertrand
- Barts and The London, Queen Mary University of London School of Medicine and Dentistry, William Harvey Research Institute, London, UK
| | - Suzanne E Eldridge
- Barts and The London, Queen Mary University of London School of Medicine and Dentistry, William Harvey Research Institute, London, UK
| | - Francesco Dell'Accio
- Barts and The London, Queen Mary University of London School of Medicine and Dentistry, William Harvey Research Institute, London, UK.
| |
Collapse
|
100
|
Weng T, Xie Y, Yi L, Huang J, Luo F, Du X, Chen L, Liu C, Chen D, Chen L. Loss of Vhl in cartilage accelerated the progression of age-associated and surgically induced murine osteoarthritis. Osteoarthritis Cartilage 2014; 22:1197-205. [PMID: 24999110 PMCID: PMC4150692 DOI: 10.1016/j.joca.2014.06.031] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 06/05/2014] [Accepted: 06/25/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the role of Vhl in maintaining the integrity of articular cartilage and in the development of experimental osteoarthritis (OA). METHOD Histology of articular cartilage and subchondral bone in both Vhl cKO and WT mice were analyzed by histopathology and micro-CT. Articular cartilage destruction and proteoglycan loss were scored in aged (12-month-old) mice as well as in mice with surgically induced OA. Apoptosis of cartilage in age-related and surgically induced OA was detected with TUNEL assay. Expressions of von Hippel-Lindau (VHL), Fas, LC-3, HIF-1α, HIF-2α, p-mTOR and MMP-13 in the knee joints were analyzed by immunostaining. RESULTS No gross differences in cartilage were observed between Vhl cKO and WT mice at age 4 months. However, Vhl cKO mice displayed accelerated age-associated spontaneous OA and surgically induced OA. Cartilage destruction and proteoglycan loss were observed in the absence of Vhl. In addition, inactivation of Vhl resulted in up-regulation of HIF-2α and increased chondrocyte apoptosis and decreased expression of autophagy during OA development. Immunohistochemical staining also showed that Vhl deficiency led to increased expression of Fas, p-mTOR and MMP-13, and those genes were associated with cell apoptosis, autophagy and cartilage matrix breakdown, respectively. CONCLUSION Loss of Vhl in adult articular cartilage is associated with earlier dysregulation of cartilage homeostasis, characterized by an increased chondrocyte apoptosis, compromised chondrocyte autophagy, and an accelerated age-related and surgery-induced OA development. These results highlight the novel role of Vhl in maintaining joint homeostasis and OA development.
Collapse
Affiliation(s)
- T Weng
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Y Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - L Yi
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - J Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - F Luo
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - X Du
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - L Chen
- Department of Orthopedics, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - C Liu
- Department of Rheumatic Disease, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - D Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - L Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| |
Collapse
|