101
|
Circular RNA RHOT1 Regulates miR-142-5p/CCND1 to Participate in Chondrocyte Autophagy and Proliferation in Osteoarthritis. J Immunol Res 2022; 2022:4370873. [PMID: 35300071 PMCID: PMC8923789 DOI: 10.1155/2022/4370873] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/08/2022] [Accepted: 01/20/2022] [Indexed: 12/13/2022] Open
Abstract
Background Osteoarthritis (OA) serves as one of the most prevalent types of joint disorders and is a leading cause of symptoms of stiffness, swelling, and arthralgia. Circular RNAs (circRNAs) have been reported to participate in various cellular processes by competing with microRNAs. Meanwhile, cyclinD1 (CCND1) is a key cell cycle regulatory protein and plays a crucial role in OA progression. Nevertheless, the function of circRHOT1 in the modulation of OA progression is still obscure. Here, we explored the effect of circRHOT1 on autophagy and extracellular matrix (ECM) in OA. Methods The expression of circRHOT1 and autophagy markers was detected in OA samples. The effect of circRHOT1 on OA was analyzed in the OA rat model. The function of circRHOT1 in the regulation of OA was assessed by CCK-8, colony formation, flow cytometry analysis, quantitative real-time PCR, Western blot analysis, and luciferase reporter gene assay in chondrocytes. Results We observed that autophagy markers, including LC3 and beclin1, were repressed in clinical OA samples. The expression of circRHOT1 and CCND1 was induced but the miR-142-5p expression was reduced in clinical OA samples. The miR-142-5p expression was negatively correlated with circRHOT1 and CCND1, and the circRHOT1 expression was positively associated with CCND1 in clinical OA samples. Meanwhile, the apoptosis was induced in OA rats but the depletion of circRHOT1 could block the phenotype in the rats. The articular cartilage degeneration was promoted in OA rats, while the knockdown of circRHOT1 repressed the degeneration. The serum levels of CTX-II and COMP were increased in OA rats, and the silencing of circRHOT1 downregulated levels of these proteins. In addition, the expression of collagen II and aggrecan was promoted by the depletion of circRHOT1 in the OA rats. Significantly, the expression of LC3 and beclin1 was suppressed in OA rats, in which the knockdown of circRHOT1 could reverse the effect. Moreover, the depletion of circRHOT1 repressed the cell viability and proliferation but induced apoptosis of chondrocytes. The expression levels of LC3, beclin1, collagen II, and aggrecan were induced by circRHOT1 knockdown. Mechanically, circRHOT1 was able to enhance the CCND1 expression by sponging miR-142-5p in chondrocytes. The overexpression of CCND1 or the inhibition of miR-142-5p reversed circRHOT1 depletion-mediated chondrocyte phenotypes. Conclusions Circular RNA RHOT1 enhances the CCND1 expression by sponging miR-142-5p to inhibit chondrocyte autophagy and promote chondrocyte proliferation in osteoarthritis. Our findings provided a promising therapeutic target for OA.
Collapse
|
102
|
Ait Eldjoudi D, Cordero Barreal A, Gonzalez-Rodríguez M, Ruiz-Fernández C, Farrag Y, Farrag M, Lago F, Capuozzo M, Gonzalez-Gay MA, Mera Varela A, Pino J, Gualillo O. Leptin in Osteoarthritis and Rheumatoid Arthritis: Player or Bystander? Int J Mol Sci 2022; 23:ijms23052859. [PMID: 35270000 PMCID: PMC8911522 DOI: 10.3390/ijms23052859] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/10/2022] Open
Abstract
White adipose tissue (WAT) is a specialized tissue whose main function is lipid synthesis and triglyceride storage. It is now considered as an active organ secreting a plethora of hormones and cytokines namely adipokines. Discovered in 1994, leptin has emerged as a key molecule with pleiotropic functions. It is primarily recognized for its role in regulating energy homeostasis and food intake. Currently, further evidence suggests its potent role in reproduction, glucose metabolism, hematopoiesis, and interaction with the immune system. It is implicated in both innate and adaptive immunity, and it is reported to contribute, with other adipokines, in the cross-talking networks involved in the pathogenesis of chronic inflammation and immune-related diseases of the musculo-skeletal system such as osteoarthritis (OA) and rheumatoid arthritis (RA). In this review, we summarize the most recent findings concerning the involvement of leptin in immunity and inflammatory responses in OA and RA.
Collapse
Affiliation(s)
- Djedjiga Ait Eldjoudi
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
| | - Alfonso Cordero Barreal
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
- International PhD School, University of Santiago de Compostela (EDIUS), 15706 Santiago de Compostela, Spain
| | - María Gonzalez-Rodríguez
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
- International PhD School, University of Santiago de Compostela (EDIUS), 15706 Santiago de Compostela, Spain
| | - Clara Ruiz-Fernández
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
- International PhD School, University of Santiago de Compostela (EDIUS), 15706 Santiago de Compostela, Spain
| | - Yousof Farrag
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
| | - Mariam Farrag
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
| | - Francisca Lago
- Molecular and Cellular Cardiology Group, SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 7, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain;
| | - Maurizio Capuozzo
- National Health Service, Local Health Authority ASL 3 Napoli Sud, Department of Pharmacy, Ercolano, 80056 Naples, Italy;
| | - Miguel Angel Gonzalez-Gay
- Hospital Universitario Marqués de Valdecilla, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, University of Cantabria, Avenida de Valdecilla s/n, 39011 Santander, Spain;
| | - Antonio Mera Varela
- SERGAS, Servizo Galego de Saude, Santiago University Clinical Hospital, Division of Rheumatology, 15706 Santiago de Compostela, Spain;
| | - Jesús Pino
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain; (D.A.E.); (A.C.B.); (M.G.-R.); (C.R.-F.); (Y.F.); (M.F.); (J.P.)
- Correspondence:
| |
Collapse
|
103
|
Carpintero-Fernández P, Varela-Eirín M, García-Yuste A, López-Díaz I, Caeiro JR, Mayán MD. Osteoarthritis: Mechanistic Insights, Senescence, and Novel Therapeutic Opportunities. Bioelectricity 2022; 4:39-47. [PMID: 39355566 PMCID: PMC11441363 DOI: 10.1089/bioe.2021.0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Osteoarthritis (OA) is the most common joint disease. In the last years, the research community has focused on understanding the molecular mechanisms that led to the pathogenesis of the disease, trying to identify different molecular and clinical phenotypes along with the discovery of new therapeutic opportunities. Different types of cell-to-cell communication mechanisms have been proposed to contribute to OA progression, including mechanisms mediated by connexin43 (Cx43) channels or by small extracellular vesicles. Furthermore, changes in the chondrocyte phenotype such as cellular senescence have been proposed as new contributors of the OA progression, changing the paradigm of the disease. The use of different drugs able to restore chondrocyte phenotype, to reduce cellular senescence and senescence-associated secretory phenotype components, and to modulate ion channel activity or Cx43 appears to be promising therapeutic strategies for the different types of OA. In this review, we aim to summarize the current knowledge in OA phenotypes related with aging and tissue damage and the new therapeutic opportunities currently available.
Collapse
Affiliation(s)
- Paula Carpintero-Fernández
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Marta Varela-Eirín
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
- European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Alejandro García-Yuste
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - Iñaki López-Díaz
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - José Ramón Caeiro
- Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| |
Collapse
|
104
|
Liu D, Cai ZJ, Yang YT, Lu WH, Pan LY, Xiao WF, Li YS. Mitochondrial quality control in cartilage damage and osteoarthritis: new insights and potential therapeutic targets. Osteoarthritis Cartilage 2022; 30:395-405. [PMID: 34715366 DOI: 10.1016/j.joca.2021.10.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is a multifactorial arthritic disease of weight-bearing joints concomitant with chronic and intolerable pain, loss of locomotion and impaired quality of life in the elderly population. Although the prevalence of OA increases with age, its specific mechanisms have not been elucidated and effective therapeutic disease-modifying drugs have not been developed. As essential organelles in chondrocytes, mitochondria supply energy and play vital roles in cellular metabolism, proliferation and apoptosis. Mitochondrial quality control (MQC) is the key mechanism to coordinate various mitochondrial biofunctions, primarily through mitochondrial biogenesis, dynamics, autophagy and the newly discovered mitocytosis. An increasing number of studies have revealed that a loss of MQC homeostasis contributes to the cartilage damage during the occurrence and development of OA. Several master MQC-associated signaling pathways and regulators exert chondroprotective roles in OA, while cartilage damage-related molecular mechanisms have been partially identified. In this review, we summarized known mechanisms mediated by dysregulated MQC in the pathogenesis of OA and latent bioactive ingredients and drugs for the prevention and treatment of OA through the maintenance of MQC.
Collapse
Affiliation(s)
- D Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Z-J Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Y-T Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - W-H Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - L-Y Pan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - W-F Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Y-S Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
105
|
Hodgeson S, O’Brien S, Simkin J, Plakotaris E, McCarthy C, Dasa V, Marrero L. Differences in synovial fibrosis relative to range of motion in knee osteoarthritis patients. J Orthop Res 2022; 40:584-594. [PMID: 33913554 PMCID: PMC8553814 DOI: 10.1002/jor.25061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 02/04/2023]
Abstract
This study tests if differences exist in the severity of synovial fibrosis between patients undergoing total knee arthroplasty (TKA) for osteoarthritis (OA) to help explain disparate deficits in pre- and postoperative range of motion (ROM) between patient groups. 117 knee OA patients were grouped by women (n = 74) and men (n = 43) or those who self-reported as Black (n = 48) or White (n = 69). ROM was measured pre- and post-TKA. Condyles and synovium collected during TKA were scored histologically for OA severity and synovitis. Fibrosis was measured from picrosirius-stained sections of the synovium. Data were analyzed using Mann-Whitney, parametric, and Spearman's rho tests with alpha at 0.05. We found no significant differences between patient age, BMI, radiographic scores, or deformity type when grouped by sex or race, or between metrics or OA severity when grouped by sex. Notably, higher synovitis was measured in women (p = .039) than men. White patients had greater ROM before (p = 0.46) and after surgery (p = .021) relative to Black patients. Fibrosis, but not OA severity and synovitis scores, for the total patient sample negatively correlated with preoperative (r s = -0.330; p = .0003) but not postoperative (rs = -0.032; p = .7627) ROM. Black patients manifested more fibrosis than White patients (p = <.0001), without significant differences between sexes. Statement of Clinical Significance: Coupled with histological scoring, measuring perioperative differences in synovial fibrosis against ROM may refine OA classification and justify the in-depth preoperative assessment of the knee as a whole. Such individualized analyses could guide personalized strategies to relieve symptomatic OA when TKA is not readily accessible and promote equitable TKA outcomes.
Collapse
Affiliation(s)
- Sydney Hodgeson
- Louisiana State University Health Sciences Center, School of Medicine, New Orleans, LA, United States
| | - Sarah O’Brien
- Louisiana State University Health Sciences Center, Morphology and Imaging Core, New Orleans, LA, United States
| | - Jennifer Simkin
- Louisiana State University Health Sciences Center, Department of Orthopaedics, New Orleans, LA, United States
| | - Elena Plakotaris
- Louisiana State University Health Sciences Center, Morphology and Imaging Core, New Orleans, LA, United States
| | - Christina McCarthy
- Louisiana State University Health Sciences Center, Department of Orthopaedics, New Orleans, LA, United States
| | - Vinod Dasa
- Louisiana State University Health Sciences Center, Department of Orthopaedics, New Orleans, LA, United States
| | - Luis Marrero
- Louisiana State University Health Sciences Center, Department of Orthopaedics, New Orleans, LA, United States,Louisiana State University Health Sciences Center, Morphology and Imaging Core, New Orleans, LA, United States,Address correspondence to Dr. Luis Marrero, Louisiana State University Health Sciences Center, 533 Bolivar St., Clinical Sciences Research Bldg., Room 608, New Orleans, LA 70112, . Ph: +1-504-568-2538
| |
Collapse
|
106
|
Wu CJ, Liu RX, Huan SW, Tang W, Zeng YK, Zhang JC, Yang J, Li ZY, Zhou Y, Zha ZG, Zhang HT, Liu N. Senescent skeletal cells cross-talk with synovial cells plays a key role in the pathogenesis of osteoarthritis. Arthritis Res Ther 2022; 24:59. [PMID: 35227288 PMCID: PMC8883702 DOI: 10.1186/s13075-022-02747-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 02/14/2022] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) has been recognized as an age-related degenerative disease commonly seen in the elderly that affects the whole “organ” including cartilage, subchondral bone, synovium, and muscles. An increasing number of studies have suggested that the accumulation of senescent cells triggering by various stresses in the local joint contributes to the pathogenesis of age-related diseases including OA. In this review, we mainly focus on the role of the senescent skeletal cells (chondrocytes, osteoblasts, osteoclasts, osteocyte, and muscle cells) in initiating the development and progression of OA alone or through cross-talk with the macrophages/synovial cells. Accordingly, we summarize the current OA-targeted therapies based on the abovementioned theory, e.g., by eliminating senescent skeletal cells and/or inhibiting the senescence-associated secretory phenotype (SASP) that drives senescence. Furthermore, the existing animal models for the study of OA from the perspective of senescence are highlighted to fill the gap between basic research and clinical applications. Overall, in this review, we systematically assess the current understanding of cellular senescence in OA, which in turn might shed light on the stratified OA treatments.
Collapse
Affiliation(s)
- Chong-Jie Wu
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ri-Xu Liu
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Song-Wei Huan
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.,Institute of Orthopedic Diseases & The Bone and Joint Disease institute of Guangdong-Hong Kong-Macao Greater Bay Area, Jinan University, Guangzhou, 510630, China
| | - Wang Tang
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yu-Kai Zeng
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Jun-Cheng Zhang
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Jie Yang
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.,Institute of Orthopedic Diseases & The Bone and Joint Disease institute of Guangdong-Hong Kong-Macao Greater Bay Area, Jinan University, Guangzhou, 510630, China
| | - Zhen-Yan Li
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.,Institute of Orthopedic Diseases & The Bone and Joint Disease institute of Guangdong-Hong Kong-Macao Greater Bay Area, Jinan University, Guangzhou, 510630, China
| | - Ying Zhou
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China.,Institute of Orthopedic Diseases & The Bone and Joint Disease institute of Guangdong-Hong Kong-Macao Greater Bay Area, Jinan University, Guangzhou, 510630, China
| | - Huan-Tian Zhang
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China. .,Institute of Orthopedic Diseases & The Bone and Joint Disease institute of Guangdong-Hong Kong-Macao Greater Bay Area, Jinan University, Guangzhou, 510630, China.
| | - Ning Liu
- Department of Bone and Joint Surgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China. .,Institute of Orthopedic Diseases & The Bone and Joint Disease institute of Guangdong-Hong Kong-Macao Greater Bay Area, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
107
|
Xu F, Hu QF, Li J, Shi CJ, Luo JW, Tian WC, Pan LW. SOX4-activated lncRNA MCM3AP-AS1 aggravates osteoarthritis progression by modulating miR-149-5p/Notch1 signaling. Cytokine 2022; 152:155805. [PMID: 35202986 DOI: 10.1016/j.cyto.2022.155805] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/04/2022] [Accepted: 01/09/2022] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To clarify the expression and underlying network of long non-coding RNA (lncRNA) MCM3AP-AS1 in osteoarthritis (OA). METHODS Human articular cartilage samples, OA model rats and IL-1β-treated C28/I2 cells were used in this study. The expression changes of genes and proteins were assessed by real-time quantitative PCR (qRT-PCR) and western blot. Cell viability, apoptosis, autophagy and extracellular matrix (ECM) degradation were assessed by Cell Counting Kit-8 (CCK-8), immunohistochemistry (IHC), flow cytometry, immunofluorescence and western blot assays, respectively. Molecule interactions were validated by dual luciferase and Chromatin immunoprecipitation (ChIP) assays. H&E staining was used to detect the pathological changes of cartilage. RESULTS MCM3AP-AS1 was upregulated in OA patients and IL-1β-induced chondrocytes. Knockdown of MCM3AP-AS1 enhanced autophagy, while alleviated ECM degradation and cartilage injury. Mechanistically, overexpression of SOX4 boosted the transcription of MCM3AP-AS1. Moreover, MCM3AP-AS1 functioned as a molecular sponge or epigenetic regulator of miR-149-5p to facilitate Notch1 expression. Functional rescue experiments showed that either inhibition of miR-149-5p nor ectopic expression of Notch1 dramatically weakened the biological impacts of MCM3AP-AS1 silencing. CONCLUSION These finding demonstrated that SOX4-activated MCM3AP-AS1 aggravated OA progression by modulating autophagy and ECM degradation via targeting miR-149-5p/Notch1 axis. These data supported that inhibition of MCM3AP-AS1 might be a potential treatment strategy of OA.
Collapse
Affiliation(s)
- Fei Xu
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China.
| | - Qun-Fang Hu
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China
| | - Jia Li
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China
| | - Chang-Jiang Shi
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China
| | - Jin-Wei Luo
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China
| | - Wei-Chao Tian
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China
| | - Li-Wei Pan
- Department of Orthopedics, Affiliated Hospital of Chengde Medical College, Chengde 067000, Hebei Province, People's Republic of China
| |
Collapse
|
108
|
Lu Y, Chen J, Li L, Cao Y, Zhao Y, Nie X, Ding C. Hierarchical functional nanoparticles boost osteoarthritis therapy by utilizing joint-resident mesenchymal stem cells. J Nanobiotechnology 2022; 20:89. [PMID: 35183192 PMCID: PMC8858465 DOI: 10.1186/s12951-022-01297-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
Utilization of joint-resident mesenchymal stem cells (MSC) to repair articular cartilage is a promising strategy in osteoarthritis (OA) therapy but remains a considerable research challenge. Here, hierarchical targeting and microenvironment responsive peptide functionalized nanoparticles (NPs) are used to achieve cartilage repair in situ. Ultrasmall copper oxide (CuO) NPs are conjugated with type 2 collagen and MSC dual-targeting peptide (designated WPV) with a matrix metalloproteinase 2 (MMP-2)-sensitive sequence as a spacer to achieve hierarchical targeting. Guided by this peptide, WPV-CuO NPs initially penetrate cartilage and subsequently expose the inner MSC-targeted peptide to attract MSCs through MMP-2 clearance. CuO further promotes chondrogenesis of MSCs. In an anterior cruciate ligament transection rat model, intraarticular injection of WPV-CuO NPs induces significant reduction of cartilage destruction. The therapeutic mechanism involves inhibition of the PI3K/AKT/mTOR pathway, as determined via transcriptome analysis. In conclusion, a novel therapeutic strategy for OA has been successfully developed based on localized MSC recruitment and cartilage repair without transplantation of exogenous cells or growth factors.
Collapse
|
109
|
Rayson A, Boudiffa M, Naveed M, Griffin J, Dall’Ara E, Bellantuono I. Geroprotectors and Skeletal Health: Beyond the Headlines. Front Cell Dev Biol 2022; 10:682045. [PMID: 35223825 PMCID: PMC8864221 DOI: 10.3389/fcell.2022.682045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis and osteoarthritis are the most common age-related diseases of the musculoskeletal system. They are responsible for high level of healthcare use and are often associated with comorbidities. Mechanisms of ageing such as senescence, inflammation and autophagy are common drivers for both diseases and molecules targeting those mechanisms (geroprotectors) have potential to prevent both diseases and their co-morbidities. However, studies to test the efficacy of geroprotectors on bone and joints are scant. The limited studies available show promising results to prevent and reverse Osteoporosis-like disease. In contrast, the effects on the development of Osteoarthritis-like disease in ageing mice has been disappointing thus far. Here we review the literature and report novel data on the effect of geroprotectors for Osteoporosis and Osteoarthritis, we challenge the notion that extension of lifespan correlates with extension of healthspan in all tissues and we highlight the need for more thorough studies to test the effects of geroprotectors on skeletal health in ageing organisms.
Collapse
Affiliation(s)
- Alexandra Rayson
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Maya Boudiffa
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Maneeha Naveed
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Jon Griffin
- Healthy Lifespan Institute, Department of Molecular Biology and Biotechnology, The University of Sheffield, Sheffield, United Kingdom
| | - Enrico Dall’Ara
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
- Insigneo Institute for in silico Medicine, Sheffield, United Kingdom
| | - Ilaria Bellantuono
- Healthy Lifespan Institute, Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| |
Collapse
|
110
|
Zhang SL, Zhang KS, Wang JF, Wang YC, Zhang H, Tang C, Pei Z, Guan ZP. Corresponding Changes of Autophagy-Related Genes and Proteins in Different Stages of Knee Osteoarthritis: An Animal Model Study. Orthop Surg 2022; 14:595-604. [PMID: 35088942 PMCID: PMC8927001 DOI: 10.1111/os.13057] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 01/13/2023] Open
Abstract
Objective To investigate the effect of autophagy expression levels of different weight‐bearing states and different stages of osteoarthritis in animal models, as well as the corresponding mechanisms. Methods We used the male Sprague–Dawley (SD) rats (12‐week‐old, SPF) to establish the OA animal models by modified Hulth method, and grouped animal models according to the length of time after surgery and different weight‐bearing areas. RT‐qPCR was carried out for detection of autophagy‐related genes such as Atg7, Atg12, P62, etc. Western blot analysis was used to detect the expression levels of corresponding autophagy‐related proteins such as LC3B, P62, etc. T test was performed for statistical analysis to compare different groups, while the differences were deemed statistically significant with P < 0.05. Transmission electron microscopy was used to observe the autophagosome to demonstrate the level of autophagy expression and the status of the chondrocytes. Results The results of the RT‐qPCR testing showed that when the weight‐bearing cartilage of the 4‐week group (relatively mild) was compared with that of the 10‐week group (relatively severe), there were statistically significant differences in all the genes tested, in detail: Atg3 (P < 0.01), Atg7 (P < 0.01), Atg12 (P < 0.01), P62 (P < 0.0001). The expression of autophagy‐related mRNA in the 4‐week group is increased compared with that of the 10‐week group. As for the expression of proteins, Western blotting showed that in the comparison between the 4‐ and the 10‐week groups, statistically significant results include Atg12 (P < 0.01) in the non‐weight‐bearing area, with decreased autophagy in the 10‐week group compared with that of the 4‐week group, while expression of LC3B (P < 0.05) protein was significantly higher in the 4‐week group than in the control in the non‐weight‐bearing area. The expression of LC3B (P < 0.0001) and P62 (P < 0.05) in the 10‐week group were higher than that of the control. Transmission electron microscope showed that autophagy in the weight‐bearing area is stronger than that in the non‐weight‐bearing area, and autophagy in the 4‐week group is stronger than in the 10‐week group for the weight‐bearing area. Conclusions The expression of autophagy varies during different stages of osteoarthritis, in which the autophagy is stronger in the early stage of osteoarthritis, and gradually decreases with the progression of the disease. Autophagy in different weight‐bearing areas may also be different.
Collapse
Affiliation(s)
- Shao-Long Zhang
- Peking University Shougang Hospital, Beijing, China.,Peking University Health Science Center, Beijing, China.,Civil Aviation General Hospital, Beijing, China
| | - Ke-Shi Zhang
- Peking University Shougang Hospital, Beijing, China
| | - Jun-Feng Wang
- Peking University International Hospital, Beijing, China
| | - Yi-Chuan Wang
- Peking University Shougang Hospital, Beijing, China.,Peking University Health Science Center, Beijing, China
| | - Hui Zhang
- Peking University Shougang Hospital, Beijing, China
| | - Chong Tang
- Peking University Shougang Hospital, Beijing, China
| | - Zheng Pei
- Peking University Shougang Hospital, Beijing, China
| | | |
Collapse
|
111
|
Cartilage degeneration is associated with activation of the PI3K/AKT signaling pathway in a growing rat experimental model of developmental trochlear dysplasia. J Adv Res 2022; 35:109-116. [PMID: 35003796 PMCID: PMC8721235 DOI: 10.1016/j.jare.2021.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 01/25/2023] Open
Abstract
Established a new experimental rat model of the developmental trochlear dysplasia; Using the macroscopic morphological and micro-CT to assess trochlear dysplasia; Using Histological staining to investigate the cartilage degradation of the model; Investigated the relationship of the PI3K/AKT signaling pathway with trochlear dysplasia cartilage degeneration; Using immunohistochemistry and qPCR to investigate the PI3K/AKT and the marker of the cartilage degeneration.
Introduction Trochlear dysplasia is a commonly encountered lower extremity deformity in humans. However, the molecular mechanism of cartilage degeneration in trochlear dysplasia is unclear thus far. Objectives The PI3K/AKT signaling pathway is known to be important for regulating the pathophysiology of cartilage degeneration. The aim of this study was to investigate the relationship of the PI3K/AKT signaling pathway with trochlear dysplasia cartilage degeneration. Methods In total, 120 female Sprague-Dawley rats (4 weeks of age) were randomly separated into control and experimental groups. Distal femurs were isolated from the experimental group at 4, 8, and 12 weeks after surgery; they were isolated from the control group at the same time points. Micro-computed tomography and histological examination were performed to investigate trochlear anatomy and changes in trochlear cartilage. Subsequently, expression patterns of PI3K/AKT, TGFβ1, and ADAMTS-4 in cartilage were investigated by immunohistochemistry and quantitative polymerase chain reaction. Results In the experimental group, the trochlear dysplasia model was successfully established at 8 weeks after surgery. Moreover, cartilage degeneration was observed beginning at 8 weeks after surgery, with higher protein and mRNA expression levels of PI3K/AKT, TGFβ1, and ADAMTS-4, relative to the control group. Conclusion Patellar instability might lead to trochlear dysplasia in growing rats. Moreover, trochlear dysplasia may cause patellofemoral osteoarthritis; cartilage degeneration in trochlear dysplasia might be associated with activation of the PI3K/AKT signaling pathway. These results provide insights regarding the high incidence of osteoarthritis in patients with trochlear dysplasia. However, more research is needed to clarify the underlying mechanisms.
Collapse
|
112
|
Yang L, Zhang Z, Wang D, Jiang Y, Liu Y. Targeting mTOR Signaling in Type 2 Diabetes Mellitus and Diabetes Complications. Curr Drug Targets 2022; 23:692-710. [PMID: 35021971 DOI: 10.2174/1389450123666220111115528] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 11/22/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a pivotal regulator of cell metabolism and growth. In the form of two different multi-protein complexes, mTORC1 and mTORC2, mTOR integrates cellular energy, nutrient and hormonal signals to regulate cellular metabolic homeostasis. In type 2 diabetes mellitus (T2DM) aberrant mTOR signaling underlies its pathological conditions and end-organ complications. Substantial evidence suggests that two mTOR-mediated signaling schemes, mTORC1-p70S6 kinase 1 (S6K1) and mTORC2-protein kinase B (AKT), play a critical role in insulin sensitivity and that their dysfunction contributes to development of T2DM. This review summaries our current understanding of the role of mTOR signaling in T2DM and its associated complications, as well as the potential use of mTOR inhibitors in treatment of T2DM.
Collapse
Affiliation(s)
- Lin Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhixin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Doudou Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Ying Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
113
|
Kuwahara M, Akasaki Y, Kurakazu I, Sueishi T, Toya M, Uchida T, Tsutsui T, Hirose R, Tsushima H, Teramura T, Nakashima Y. C10orf10/DEPP activates mitochondrial autophagy and maintains chondrocyte viability in the pathogenesis of osteoarthritis. FASEB J 2022; 36:e22145. [PMID: 34997944 DOI: 10.1096/fj.202100896r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA), the most prevalent joint disease, is characterized by the progressive loss of articular cartilage. Autophagy, a lysosomal degradation pathway, maintains cellular homeostasis, and autophagic dysfunction in chondrocytes is a hallmark of OA pathogenesis. However, the cause of autophagic dysfunction in OA chondrocytes remains incompletely understood. Recent studies have reported that decidual protein induced by progesterone (C10orf10/DEPP) positively regulates autophagic functions. In this study, we found that DEPP was involved in mitochondrial autophagic functions of chondrocytes, as well as in OA pathogenesis. DEPP expression decreased in human OA chondrocytes in the absence or presence of pro-inflammatory cytokines, and was induced by starvation, hydrogen peroxide (H2 O2 ), and hypoxia (cobalt chloride). For functional studies, DEPP knockdown decreased autophagic flux induced by H2 O2 , whereas DEPP overexpression increased autophagic flux and maintained cell viability following H2 O2 treatment. DEPP was downregulated by knockdown of forkhead box class O (FOXO) transcription factors and modulated the autophagic function regulated by FOXO3. In an OA mouse model by destabilization of the medial meniscus, DEPP-knockout mice exacerbated the progression of cartilage degradation with TUNEL-positive cells, and chondrocytes isolated from knockout mice were decreased autophagic flux and increased cell death following H2 O2 treatment. Subcellular fractionation analysis revealed that mitochondria-located DEPP activated mitochondrial autophagy via BCL2 interacting protein 3. Taken together, our data demonstrate that DEPP is a major stress-inducible gene involved in the activation of mitochondrial autophagy in chondrocytes, and maintains chondrocyte viability during OA pathogenesis. DEPP represents a potential therapeutic target for enhancing autophagy in patients with OA.
Collapse
Affiliation(s)
- Masanari Kuwahara
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Yukio Akasaki
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Ichiro Kurakazu
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Takuya Sueishi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Taisuke Uchida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Tomoaki Tsutsui
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Ryota Hirose
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Hidetoshi Tsushima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka-sayama, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka City, Japan
| |
Collapse
|
114
|
Bauza‐Mayol G, Quintela M, Brozovich A, Hopson M, Shaikh S, Cabrera F, Shi A, Niclot FB, Paradiso F, Combellack E, Jovic T, Rees P, Tasciotti E, Francis LW, Mcculloch P, Taraballi F. Biomimetic Scaffolds Modulate the Posttraumatic Inflammatory Response in Articular Cartilage Contributing to Enhanced Neoformation of Cartilaginous Tissue In Vivo. Adv Healthc Mater 2022; 11:e2101127. [PMID: 34662505 PMCID: PMC11469755 DOI: 10.1002/adhm.202101127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/28/2021] [Indexed: 12/13/2022]
Abstract
Focal chondral lesions of the knee are the most frequent type of trauma in younger patients and are associated with a high risk of developing early posttraumatic osteoarthritis. The only current clinical solutions include microfracture, osteochondral grafting, and autologous chondrocyte implantation. Cartilage tissue engineering based on biomimetic scaffolds has become an appealing strategy to repair cartilage defects. Here, a chondrogenic collagen-chondroitin sulfate scaffold is tested in an orthotopic Lapine in vivo model to understand the beneficial effects of the immunomodulatory biomaterial on the full chondral defect. Using a combination of noninvasive imaging techniques, histological and whole transcriptome analysis, the scaffolds are shown to enhance the formation of cartilaginous tissue and suppression of host cartilage degeneration, while also supporting tissue integration and increased tissue regeneration over a 12 weeks recovery period. The results presented suggest that biomimetic materials could be a clinical solution for cartilage tissue repair, due to their ability to modulate the immune environment in favor of regenerative processes and suppression of cartilage degeneration.
Collapse
Affiliation(s)
- Guillermo Bauza‐Mayol
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
- Reproductive Biology and Gynaecological Oncology GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Marcos Quintela
- Reproductive Biology and Gynaecological Oncology GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Ava Brozovich
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
- Texas A&M College of MedicineBryanTX77807USA
| | - Michael Hopson
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Shazad Shaikh
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Fernando Cabrera
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Aaron Shi
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Federica Banche Niclot
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Polytechnic of TurinDepartment of Applied Science and TechnologyCorso Duca degli Abruzzi 24Torino10129Italy
| | - Francesca Paradiso
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
- Reproductive Biology and Gynaecological Oncology GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Emman Combellack
- Reconstructive Surgery and Regenerative Medicine Research GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Tom Jovic
- Reconstructive Surgery and Regenerative Medicine Research GroupSwansea University Medical SchoolSingleton ParkSwanseaSA2 8PPUK
| | - Paul Rees
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | | | - Lewis W. Francis
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
| | - Patrick Mcculloch
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| | - Francesca Taraballi
- Center for Musculoskeletal RegenerationHouston Methodist Research Institute6670 Bertner Ave.HoustonTX77030USA
- Orthopedics & Sports MedicineHouston Methodist Hospital6550 Fannin St.HoustonTX77030USA
| |
Collapse
|
115
|
Abstract
PURPOSE OF REVIEW Translation of genetic information encoded within mRNA molecules by ribosomes into proteins is a key part of the central dogma of molecular biology. Despite the central position of the ribosome in the translation of proteins, and considering the major proteomic changes that occur in the joint during osteoarthritis development and progression, the ribosome has received very limited attention as driver of osteoarthritis pathogenesis. RECENT FINDINGS We provide an overview of the limited literature regarding this developing topic for the osteoarthritis field. Recent key findings that connect ribosome biogenesis and activity with osteoarthritis include: ribosomal RNA transcription, processing and maturation, ribosomal protein expression, protein translation capacity and preferential translation. SUMMARY The ribosome as the central cellular protein synthesis hub is largely neglected in osteoarthritis research. Findings included in this review reveal that in osteoarthritis, ribosome aberrations have been found from early-stage ribosome biogenesis, through ribosome build-up and maturation, up to preferential translation. Classically, osteoarthritis has been explained as an imbalance between joint tissue anabolism and catabolism. We postulate that osteoarthritis can be interpreted as an acquired ribosomopathy. This hypothesis fine-tunes the dogmatic anabolism/katabolism point-of-view, and may provide novel molecular opportunities for the development of osteoarthritis disease-modifying treatments.
Collapse
Affiliation(s)
- Guus G.H. van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
| | - Marjolein M.J. Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
| | - Mandy J. Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Tim J.M. Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
116
|
Arias C, Salazar LA. Autophagy and Polyphenols in Osteoarthritis: A Focus on Epigenetic Regulation. Int J Mol Sci 2021; 23:ijms23010421. [PMID: 35008847 PMCID: PMC8745146 DOI: 10.3390/ijms23010421] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an intracellular mechanism that maintains cellular homeostasis in different tissues. This process declines in cartilage due to aging, which is correlated with osteoarthritis (OA), a multifactorial and degenerative joint disease. Several studies show that microRNAs regulate different steps of autophagy but only a few of them participate in OA. Therefore, epigenetic modifications could represent a therapeutic opportunity during the development of OA. Besides, polyphenols are bioactive components with great potential to counteract diseases, which could reverse altered epigenetic regulation and modify autophagy in cartilage. This review aims to analyze epigenetic mechanisms that are currently associated with autophagy in OA, and to evaluate whether polyphenols are used to reverse the epigenetic alterations generated by aging in the autophagy pathway.
Collapse
Affiliation(s)
- Consuelo Arias
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile;
- Department of Preclinical Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile;
- Interuniversity Center for Healthy Aging (ICHA), Universidad de La Frontera, Temuco 4811230, Chile
- Correspondence: ; Tel.: +56-45-259-6724
| |
Collapse
|
117
|
Zhu X, Dai S, Xia B, Gong J, Ma B. Activation of the alpha 7 nicotinic acetylcholine receptor mitigates osteoarthritis progression by inhibiting NF-κB/NLRP3 inflammasome activation and enhancing autophagy. PLoS One 2021; 16:e0256507. [PMID: 34941874 PMCID: PMC8699641 DOI: 10.1371/journal.pone.0256507] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/09/2021] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease characterized by cartilage degradation. Alpha 7 nicotinic acetylcholine receptor (α7nAChR) is associated with inflammatory and metabolic responses in OA. However, the mechanisms underlying the pathological process of OA remain unclear. The aim of the present study was to examine the role and mechanisms of α7nAChR-mediated autophagy and anti-inflammatory response in chondroprotection. Monosodium iodoacetate (MIA)-induced Wistar rat OA model was used to assess the in vivo effects of the ɑ7nAChR agonist (PNU-282987). The histopathological characteristics of OA were evaluated by immunohistochemistry (IHC), and the levels of autophagy markers were determined by western blotting and transmission electron microscopy. The anti-inflammatory effect of the ɑ7nAChR agonist was assessed by IHC, quantitative real-time polymerase chain reaction, and western blotting. Parallel experiments to determine the molecular mechanisms through which the ɑ7nAChR agonist prevents OA were performed using interleukin-1β (IL-1β)-treated chondrocytes. Our results showed that PNU-282987 reduced cartilage degeneration and matrix metalloproteinase (MMP)-1 and MMP-13 expressions. Activating α7nAChR with PNU-282987 significantly promoted MIA/IL-1β-induced chondrocyte autophagy, as demonstrated by the increase in LC3-II/LC3-I ratio, Beclin-1 levels, and autophagosome number. Furthermore, treating chondrocyte with ULK1 siRNA attenuated the PNU282987-induced enhancement of LC3-II/LC3-I ratio and Beclin-1 level. Additionally, PNU282987 suppressed NF-κB/NLRP3 inflammasome activation by inhibiting the ROS/TXNIP pathway and suppressed tumor necrosis factor-ɑ and IL-1β secretion in MIA/IL-1β-treated chondrocytes. Our results demonstrate that the activation of α7nAChR promotes chondrocyte autophagy and attenuates inflammation to mitigate OA progression, providing a novel target for the treatment of OA.
Collapse
Affiliation(s)
- Xianjie Zhu
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Shiyou Dai
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Baohua Xia
- Department of Clinical Skills Training Center, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Jianbao Gong
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Bingzheng Ma
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, Shandong, China
- * E-mail:
| |
Collapse
|
118
|
Dai Y, Jian C, Wang X, Dai X. Comprehensive expression profiles of mRNAs, lncRNAs and miRNAs in Kashin-Beck disease identified by RNA-sequencing. Mol Omics 2021; 18:154-166. [PMID: 34913457 DOI: 10.1039/d1mo00370d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Kashin-Beck disease (KBD) is a chronic, endemic and deforming osteochondropathy, whose basic pathological alterations include apoptosis and necrosis of chondrocytes in articular cartilage and growth plates and imbalanced extracellular matrix metabolism. Numerous studies have reported that long noncoding RNAs (lncRNAs) and microRNA (miRNAs) are aberrantly expressed in KBD. Our study was comprised of 5 KBD patients and 5 healthy individuals and we compared the expression profiles of mRNAs, lncRNAs and miRNAs through RNA-sequencing (RNA-seq). Bioinformatic analysis of GO and KEGG was employed to conduct functional annotation and pathway enriched analysis. In total, 3194 mRNAs, 4103 lncRNAs and 1550 miRNAs were detected to be differentially expressed by RNA-seq (P < 0.05; |log2FC| ≥1). The lysosome pathway, Wnt signaling pathway, TNF signaling pathway, endocytosis and mTOR signaling pathway were identified to be involved in the KBD development according to the result of the KEGG analysis. In addition, a ceRNA network based on lncRNA-miRNA-mRNA was constructed to probe the intricate regulatory mechanism and interaction between transcripts, which was visualized using the Cytoscape software. The ce-lncRNAs of four aberrantly expressed genes, FOSB, EGR3, BCAM and SOX6, were determined through the network. Among the identified DElncRNAs, we selected 8 differentially expressed lncRNAs to confirm the reliability of RNA-seq data by qRT-PCR in 11 KBD patients and 11 healthy individuals. We aimed to provide a comprehensive understanding ofmRNA, lncRNA and miRNA alterations between KBD patients and healthy individuals, and meanwhile reveal several potential causative molecular and signaling pathways involved in KBD.
Collapse
Affiliation(s)
- Yu Dai
- School of Public Health, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Can Jian
- School of Public Health, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Xiaofeng Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Xiaoxia Dai
- School of Public Health, Xi'an Jiaotong University Health Science Center, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, China.,Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
119
|
Intra-Articular Drug Delivery for Osteoarthritis Treatment. Pharmaceutics 2021; 13:pharmaceutics13122166. [PMID: 34959445 PMCID: PMC8703898 DOI: 10.3390/pharmaceutics13122166] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease affecting millions of people worldwide. Currently, clinical nonsurgical treatments of OA are only limited to pain relief, anti-inflammation, and viscosupplementation. Developing disease-modifying OA drugs (DMOADs) is highly demanded for the efficient treatment of OA. As OA is a local disease, intra-articular (IA) injection directly delivers drugs to synovial joints, resulting in high-concentration drugs in the joint and reduced side effects, accompanied with traditional oral or topical administrations. However, the injected drugs are rapidly cleaved. By properly designing the drug delivery systems, prolonged retention time and targeting could be obtained. In this review, we summarize the drugs investigated for OA treatment and recent advances in the IA drug delivery systems, including micro- and nano-particles, liposomes, and hydrogels, hoping to provide some information for designing the IA injected formulations.
Collapse
|
120
|
Zeng J, Zhang Z, Liao Q, Lu Q, Liu J, Yuan L, Liu G. CircPan3 Promotes the Ghrelin System and Chondrocyte Autophagy by Sponging miR-667-5p During Rat Osteoarthritis Pathogenesis. Front Cell Dev Biol 2021; 9:719898. [PMID: 34869311 PMCID: PMC8640465 DOI: 10.3389/fcell.2021.719898] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
This study aimed to investigate the potential roles of circRNAs in regulating osteoarthritis (OA)-related ghrelin synthesis, autophagy induction, and the relevant molecular mechanisms. Results showed that Col2a1, Acan, ghrelin, and autophagy-related markers expression were downregulated, while matrix metalloproteinase 13 (MMP13) and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) expressions increased in both IL-1β-induced rat chondrocytes and cartilage tissues of OA rats. A total of 130 circRNAs and 731 mRNAs were differentially expressed in IL-1β-induced rat chondrocytes. Among them, we found that circPan3 expression was significantly decreased in both cellular and animal OA models. CircPan3 directly targeted miR-667-5p. CircPan3 overexpression promoted Col2a1, Acan, ghrelin, beclin 1, and LC3-II expression but reduced MMP13 and ADAMTS5 expression in rat chondrocytes, whereas overexpression of miR-667-5p exhibited opposite effects on the above markers. Furthermore, we found that miR-667-5p bound directly to the 3′-UTR sequence of ghrelin gene. Moreover, the circPan3-induced alterations in chondrocytes were antagonized by miR-667-5p overexpression. Taken together, our findings demonstrate that circPan3 promotes ghrelin synthesis and chondrocyte autophagy via targeting miR-667-5p, protecting against OA injury. This study provided experimental evidence that circPan3/miR-667-5p/ghrelin axis might serve as targets of drug development for the treatment of OA.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.,Department of Rehabilitation Medicine, Nanfang University of Science and Technology Hospital, Shenzhen, China
| | - Zhenzhen Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.,Department of Rehabilitation Medicine, Hankou Hospital, Wuhan, China
| | - Qing Liao
- Department of Rehabilitation Medicine, Shunde Hospital of Southern Medical University, Southern Medical University, Foshan, China
| | - Qijin Lu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jiemei Liu
- Department of Rehabilitation Medicine, Shunde Hospital of Southern Medical University, Southern Medical University, Foshan, China
| | - Lixia Yuan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Gang Liu
- Department of Rehabilitation Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Rehabilitation Medicine, Shunde Hospital of Southern Medical University, Southern Medical University, Foshan, China
| |
Collapse
|
121
|
Nieto-Torres JL, Hansen M. Macroautophagy and aging: The impact of cellular recycling on health and longevity. Mol Aspects Med 2021; 82:101020. [PMID: 34507801 PMCID: PMC8671213 DOI: 10.1016/j.mam.2021.101020] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/11/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Aging is associated with many deleterious changes at the cellular level, including the accumulation of potentially toxic components that can have devastating effects on health. A key protective mechanism to this end is the cellular recycling process called autophagy. During autophagy, damaged or surplus cellular components are delivered to acidic vesicles called lysosomes, that secure degradation and recycling of the components. Numerous links between autophagy and aging exist. Autophagy declines with age, and increasing evidence suggests that this reduction plays important roles in both physiological aging and the development of age-associated disorders. Studies in pharmacologically and genetically manipulated model organisms indicate that defects in autophagy promote age-related diseases, and conversely, that enhancement of autophagy has beneficial effects on both healthspan and lifespan. Here, we review our current understanding of the role of autophagy in different physiological processes and their molecular links with aging and age-related diseases. We also highlight some recent advances in the field that could accelerate the development of autophagy-based therapeutic interventions.
Collapse
Affiliation(s)
- Jose L Nieto-Torres
- Sanford Burnham Prebys Medical Discovery Institute. Program of Development, Aging, and Regeneration, La Jolla, CA, USA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute. Program of Development, Aging, and Regeneration, La Jolla, CA, USA.
| |
Collapse
|
122
|
Wang XJ, Tian W, Xu WW, Lu X, Zhang YM, Li LJ, Chang F. Loss of Autophagy Causes Increased Apoptosis of Tibial Plateau Chondrocytes in Guinea Pigs with Spontaneous Osteoarthritis. Cartilage 2021; 13:796S-807S. [PMID: 34493119 PMCID: PMC8804872 DOI: 10.1177/19476035211044820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE The goal of the present study was to observe the effect of autophagy in tibial plateau chondrocytes on apoptosis in spontaneous knee osteoarthritis (OA) in guinea pigs. DESIGN Fifty 2-month-old female Hartley guinea pigs were divided into a normal group (10 animals, all euthanized after 7 months) and an OA group (40 animals, 10 of which were euthanized after 10 months). Immunohistochemistry, RT-qPCR and Western blotting were used to evaluate autophagy levels, intracellular glycogen accumulation and apoptosis in tibial plateau chondrocytes in vivo and in vitro. The remaining 30 guinea pigs in the OA group were divided into 3 groups: a rapamycin group, a normal saline group, and a 3-methyladenine (3-MA) group. Intracellular glycogen accumulation and chondrocyte apoptosis were assessed by altering the level of autophagy in chondrocytes in vivo. RESULTS When spontaneous OA occurred in guinea pigs, autophagy levels in tibial plateau chondrocytes decreased, while intracellular glycogen accumulation and the rate of chondrocyte apoptosis increased. After enhancing the level of autophagy in tibial plateau chondrocytes in guinea pigs with OA, intracellular glycogen accumulation and the rate of chondrocyte apoptosis decreased, while inhibiting autophagy had the opposite effects. CONCLUSION The results indicate that the function of autophagy in chondrocytes may at least partly involve the catabolism of glycogen. In guinea pigs with OA, the level of autophagy in tibial plateau chondrocytes decreased, and chondrocytes were unable to degrade intracellular glycogen into glucose, leading to less energy for chondrocytes and increased apoptosis.
Collapse
Affiliation(s)
- Xiao-jian Wang
- Department of Orthopaedic
Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Wei Tian
- Department of Orthopaedic
Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Wei-wei Xu
- Shanxi Key Laboratory of Kidney
Disease, Taiyuan, China
| | - Xiao Lu
- Shanxi Key Laboratory of Kidney
Disease, Taiyuan, China
| | - Yu-ming Zhang
- Department of Orthopaedic
Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Li-jun Li
- Department of Orthopaedic
Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Feng Chang
- Department of Orthopaedic
Surgery, Shanxi Provincial People’s Hospital, Taiyuan, China
| |
Collapse
|
123
|
Liu J, Fu Q, Liu S. Transcriptional Regulation Based on Network of Autophagy Identifies Key Genes and Potential Mechanisms in Human Osteoarthritis. Cartilage 2021; 13:1431S-1441S. [PMID: 32819149 PMCID: PMC8804715 DOI: 10.1177/1947603520951632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a chronic arthropathy that frequently occurs in the middle-aged and elderly population. The aim of this study was to investigate the molecular mechanism of OA based on autophagy theory. DESIGN We downloaded the gene expression profile from the Gene Expression Omnibus repository. Differentially expressed genes (DEGs) related to the keyword "autophagy" were identified using the scanGEO online analysis tool. DEGs representing the same expression trend were screened using the MATCH function. Clinical synovial specimens were collected for identification, pathological diagnosis, hematoxylin and eosin staining, and real-time polymerase chain reaction analysis. Differential expression of mRNAs in the synovial membrane tissues and chondrocyte monolayer samples from OA patients was used to identify potential OA biomarkers. Protein-protein interactions were established by the STRING website and visualized with Cytoscape. Functional and pathway enrichment analyses were performed using the Metascape database. RESULTS GABARAPL1, GABARAPL2, and ATG13 were obtained as co-expressed autogenes in the 3 data sets. They were all downregulated among OA synovial tissues compared with non-OA synovial tissues (P < 0.01). A protein-protein interaction network was constructed based on these 3 genes and included 63 genes. A functional analysis revealed that these genes were associated with autophagy-related functions. The top hub genes in the protein-protein interaction network were presented. Furthermore, 3 key modules were extracted to be core control modules. CONCLUSIONS These results offer an important molecular understanding of the key transcriptional regulatory genes and modules based on the network of potential autophagy mechanisms in human OA.
Collapse
Affiliation(s)
- Jiamei Liu
- Department of Pathology, The Shengjing
Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of
China
| | - Qin Fu
- Department of Orthopedics, The Shengjing
Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of
China
| | - Shengye Liu
- Department of Orthopedics, The Shengjing
Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of
China,Shengye Liu, Department of Orthopedics, The
Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004,
People’s Republic of China.
| |
Collapse
|
124
|
Chen L, Ni Z, Huang J, Zhang R, Zhang J, Zhang B, Kuang L, Sun X, Zhang D, Su N, Qi H, Yang J, Jin M, Luo F, Chen H, Zhou S, Du X, Ouyang J, Wang Z, Xie Y, Tan Q, Chen L. Long term usage of dexamethasone accelerating accelerates the initiation of osteoarthritis via enhancing chondrocyte apoptosis and the extracellular matrix calcification and apoptosis of chondrocytes. Int J Biol Sci 2021; 17:4140-4153. [PMID: 34803488 PMCID: PMC8579451 DOI: 10.7150/ijbs.64152] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/23/2021] [Indexed: 12/29/2022] Open
Abstract
Systemic application of glucocorticoids is an essential anti-inflammatory and immune-modulating therapy for severe inflammatory or autoimmunity conditions. However, its long-term effects on articular cartilage of patients' health need to be further investigated. In this study, we studied the effects of dexamethasone (Dex) on the homeostasis of articular cartilage and the progress of destabilization of medial meniscus (DMM)-induced osteoarthritis (OA) in adult mice. Long-term administration of Dex aggravates the proteoglycan loss of articular cartilage and drastically accelerates cartilage degeneration under surgically induced OA conditions. In addition, Dex increases calcium content in calcified cartilage layer of mice and the samples from OA patients with a history of long-term Dex treatment. Moreover, long term usage of Dex results in decrease subchondral bone mass and bone density. Further studies showed that Dex leads to calcification of extracellular matrix of chondrocytes partially through activation of AKT, as well as promotes apoptosis of chondrocytes in calcified cartilage layer. Besides, Dex weakens the stress-response autophagy with the passage of time. Taken together, our data indicate that long-term application of Dex may predispose patients to OA and or even accelerate the OA disease progression development of OA patients.
Collapse
Affiliation(s)
- Liang Chen
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China.,Department of orthopedic, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhong Ni
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Junlan Huang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Ruobin Zhang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jinfan Zhang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Zhang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Liang Kuang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xianding Sun
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Dali Zhang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Nan Su
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Huabing Qi
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fengtao Luo
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Hangang Chen
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Siru Zhou
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaolan Du
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Junjie Ouyang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zuqiang Wang
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiaoyan Tan
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
125
|
Li Z, Huang Z, Zhang H, Lu J, Tian Y, Piao S, Lin Z, Bai L. Moderate-intensity exercise alleviates pyroptosis by promoting autophagy in osteoarthritis via the P2X7/AMPK/mTOR axis. Cell Death Dis 2021; 7:346. [PMID: 34759265 PMCID: PMC8580998 DOI: 10.1038/s41420-021-00746-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/19/2021] [Accepted: 10/27/2021] [Indexed: 01/17/2023]
Abstract
Instability and excessive use of the knee joint can cause osteoarthritis (OA). Reasonable exercise can enhance the stability of the knee joint and prevent and relieve the occurrence and development of OA. As a key switch for inflammation, P2X purinoceptor 7 (P2X7) has attracted much attention in studies of OA. Exercise can regulate P2X7 expression and activation. However, the role of P2X7 in exercise-based prevention and treatment of OA is unknown. We previously showed that moderate-intensity exercise can significantly alleviate OA symptoms. Accordingly, in this study, we evaluated the effects of exercise on P2X7 expression and activation in chondrocytes. Micro-computed tomography, hematoxylin, and eosin staining, Toluidine Blue O staining, immunohistochemistry, and terminal deoxynucleotidyl transferase dUTP nick-end labeling experiments showed that P2X7 expression was lower in the moderate-intensity exercise group than in the inflammation and low- and high-intensity exercise groups. Additionally, chondrocyte death, cartilage destruction, and the degree and severity of pyroptosis were significantly reduced, whereas autophagy levels were significantly increased in the moderate-intensity exercise group. Cell Counting Kit-8 assay, lactate dehydrogenase release, flow cytometry, enzyme-linked immunosorbent assay, cell fluorescence, western blot, reverse transcription-quantitative polymerase chain reaction, and transmission electron microscopy experiments showed that moderate activation of P2X7 promoted autophagy through the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway and promoted autolysosome targeting for degradation of the inflammasome component NLRP3, thereby inhibiting pyroptosis. Additionally, the use of AMPK and mTOR activators and inhibitors indicated that the AMPK-mTOR signaling pathway, as the downstream of P2X7, played a key role in delaying the occurrence and development of OA. We propose that moderate-intensity exercise promoted chondrocyte autophagy through the P2X7/AMPK/mTOR signal axis to alleviate pyroptosis. Our findings provide novel insights into the positive and preventative effects of exercise on OA.
Collapse
Affiliation(s)
- Zihao Li
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024 China
| | - Ziyu Huang
- grid.412531.00000 0001 0701 1077Foreign Languages College, Shanghai Normal University, Shanghai, 200234 China
| | - He Zhang
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024 China
| | - Jinghan Lu
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024 China
| | - Yicheng Tian
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024 China
| | - Shang Piao
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024 China
| | - Zhiming Lin
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024 China
| | - Lunhao Bai
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 110024, China.
| |
Collapse
|
126
|
José Alcaraz M. New potential therapeutic approaches targeting synovial fibroblasts in rheumatoid arthritis. Biochem Pharmacol 2021; 194:114815. [PMID: 34715065 DOI: 10.1016/j.bcp.2021.114815] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022]
Abstract
Synovial cells play a key role in joint destruction during chronic inflammation. In particular, activated synovial fibroblasts (SFs) undergo intrinsic alterations leading to an aggressive phenotype mediating cartilage destruction and bone erosion in rheumatoid arthritis (RA). Recent research has revealed a number of targets to control arthritogenic changes in SFs. Therefore, identification of SF phenotypes, control of epigenetic changes, modulation of cellular functions, or regulation of the activity of cation channels and different signaling pathways has been investigated. Although many of these approaches have shown efficacy in vitro and in animal models of RA, further research is needed to select the most relevant targets for drug development. This review is focused on the role of SFs as a potential strategy to discover novel therapeutic targets in RA aimed at preserving joint architecture and function.
Collapse
Affiliation(s)
- María José Alcaraz
- Department of Pharmacology, University of Valencia, and Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), Polytechnic University of Valencia, University of Valencia, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| |
Collapse
|
127
|
Cao R, Yu H, Long H, Zhang H, Hao C, Shi L, Du Y, Jiao S, Guo A, Ma L, Wang Z. Low Deacetylation Degree Chitosan Oligosaccharide Protects against IL-1β Induced Inflammation and Enhances Autophagy Activity in Human Chondrocytes. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:517-531. [PMID: 34704529 DOI: 10.1080/09205063.2021.1996962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease, which can lead to joint pain, stiffness, deformity and dysfunction, that seriously affects the quality of life in patients. At present, the treatments of OA mainly include early pharmacological treatment and late joint replacement. However, current pharmacological treatment has limited efficacy and undesired side effects.Chitosan oligosaccharide (COS) is a kind of nontoxic and biodegradable oligo-saccharide, which is composed of 2-20 glucosamine or N-acetylglucosamine linked by β-1,4 glycosidic bond. Studies have shown that COS has significant biological properties like antimicrobial, anti-inflammatory, antioxidant, and anti-tumor, as well as immunoregulation ability. However, the effects of COS on OA have not been clarified. In this study, we explored the protective effects of COS with different degrees of deacetylation on chondrocytes stimulated by interleukin 1β (IL-1β) in vitro.The results showed that IL-1β inhibited cell proliferation and promoted cell apoptosis. Besides that, IL-1β increased the expression of the major chondro-degrading genes MMP13 and ADAMTS-5, while decreased the expression of COL2A and ACAN. COS with different degrees of deacetylation (HDACOS, MDACOS, LDACOS) had different effects on IL-1β induced inflammation. LDACOS had the most obvious anti-inflammatory effects to inhibit the expression of MMP13 and ADAMTS-5 while promoted the expression of COL2A and ACAN. In addition, we found that the expression of autophagy-related gene Beclin-1 was up-regulated, and the ratio of LC3-II/LC3-I was increased in the LDACOS group. Furthermore, transmission electron microscopy (TEM) analysis showed that the number of intracellular autophagosomes increased significantly with the treatment of LDACOS. Based on our research, we suggested that LDACOS could inhibit chondrocytes inflammation and promote cell autophagy, and might be a protective drug for the treatment of OA.
Collapse
Affiliation(s)
- Ruiqi Cao
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Haomiao Yu
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Huibin Long
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hongrui Zhang
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chao Hao
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lin Shi
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yuguang Du
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Siming Jiao
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Ai Guo
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lifeng Ma
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhuo Wang
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
128
|
Minton DM, Elliehausen CJ, Javors MA, Santangelo KS, Konopka AR. Rapamycin-induced hyperglycemia is associated with exacerbated age-related osteoarthritis. Arthritis Res Ther 2021; 23:253. [PMID: 34620223 PMCID: PMC8495984 DOI: 10.1186/s13075-021-02637-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The objective of this study was to determine if mechanistic target of rapamycin (mTOR) inhibition with or without AMP-activated protein kinase (AMPK) activation can protect against primary, age-related OA. DESIGN Dunkin-Hartley guinea pigs develop mild primary OA pathology by 5 months of age that progresses to moderate OA by 8 months of age. At 5 months, guinea pigs served as young control (n = 3) or were fed either a control diet (n = 8), a diet enriched with the mTOR-inhibitor rapamycin (Rap, 14 ppm, n = 8), or Rap with the AMPK-activator metformin (Rap+Met, 1000 ppm, n = 8) for 12 weeks. Knee joints were evaluated by OARSI scoring, micro-computed tomography, and immunohistochemistry. Glenohumeral articular cartilage was collected for western blotting. RESULTS Rap- and Rap+Met-treated guinea pigs displayed lower body weight than control. Rap and Rap+Met inhibited articular cartilage mTORC1 but not mTORC2 signaling. Rap+Met, but not Rap alone, stimulated AMPK. Despite lower body weight and articular cartilage mTORC1 inhibition, Rap- and Rap+Met-treated guinea pigs had greater OA severity in the medial tibial plateau due to articular cartilage structural damage and/or proteoglycan loss. Rap and Rap+Met increased plasma glucose compared to control. Plasma glucose concentration was positively correlated with proteoglycan loss, suggesting hyperglycemic stress after Rap treatment was related to worsened OA. CONCLUSIONS This is the first study to show that Rap induced increase in plasma glucose was associated with greater OA severity. Further, articular cartilage mTORC1 inhibition and bodyweight reduction by dietary Rap and Rap+Met did not appear to protect against primary OA during the prevailing hyperglycemia.
Collapse
Affiliation(s)
- Dennis M Minton
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Kinesiology, University of Illinois at Urbana-Champaign, Champaign, Illinois , USA
| | - Christian J Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Kinesiology, University of Illinois at Urbana-Champaign, Champaign, Illinois , USA
| | - Martin A Javors
- Departments of Psychiatry and Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Kelly S Santangelo
- Department of Microbiology, Immunology, Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Adam R Konopka
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.
- Department of Kinesiology, University of Illinois at Urbana-Champaign, Champaign, Illinois , USA.
- Geriatric Research, Education, and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA.
| |
Collapse
|
129
|
Ge Y, Chen Z, Fu Y, Xiao X, Xu H, Shan L, Tong P, Zhou L. Identification and validation of hub genes of synovial tissue for patients with osteoarthritis and rheumatoid arthritis. Hereditas 2021; 158:37. [PMID: 34583778 PMCID: PMC8480049 DOI: 10.1186/s41065-021-00201-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 09/01/2021] [Indexed: 12/27/2022] Open
Abstract
Background Osteoarthritis (OA) and rheumatoid arthritis (RA) were two major joint diseases with similar clinical phenotypes. This study aimed to determine the mechanistic similarities and differences between OA and RA by integrated analysis of multiple gene expression data sets. Methods Microarray data sets of OA and RA were obtained from the Gene Expression Omnibus (GEO). By integrating multiple gene data sets, specific differentially expressed genes (DEGs) were identified. The Gene Ontology (GO) functional annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and protein–protein interaction (PPI) network analysis of DEGs were conducted to determine hub genes and pathways. The “Cell Type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT)” algorithm was employed to evaluate the immune infiltration cells (IICs) profiles in OA and RA. Moreover, mouse models of RA and OA were established, and selected hub genes were verified in synovial tissues with quantitative polymerase chain reaction (qPCR). Results A total of 1116 DEGs were identified between OA and RA. GO functional enrichment analysis showed that DEGs were enriched in regulation of cell morphogenesis involved in differentiation, positive regulation of neuron differentiation, nuclear speck, RNA polymerase II transcription factor complex, protein serine/threonine kinase activity and proximal promoter sequence-specific DNA binding. KEGG pathway analysis showed that DEGs were enriched in EGFR tyrosine kinase inhibitor resistance, ubiquitin mediated proteolysis, FoxO signaling pathway and TGF-beta signaling pathway. Immune cell infiltration analysis identified 9 IICs with significantly different distributions between OA and RA samples. qPCR results showed that the expression levels of the hub genes (RPS6, RPS14, RPS25, RPL11, RPL27, SNRPE, EEF2 and RPL19) were significantly increased in OA samples compared to their counterparts in RA samples (P < 0.05). Conclusion This large-scale gene analyses provided new insights for disease-associated genes, molecular mechanisms as well as IICs profiles in OA and RA, which may offer a new direction for distinguishing diagnosis and treatment between OA and RA.
Collapse
Affiliation(s)
- Yanzhi Ge
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China
| | - Yanbin Fu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, P. R. China
| | - Xiujuan Xiao
- College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, P. R. China
| | - Haipeng Xu
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China.
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China.
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P. R. China.
| |
Collapse
|
130
|
Liao B, Guan M, Tan Q, Wang G, Zhang R, Huang J, Liu M, Chen H, Li K, Bai D, Zhu Y. Low-intensity pulsed ultrasound inhibits fibroblast-like synoviocyte proliferation and reduces synovial fibrosis by regulating Wnt/β-catenin signaling. J Orthop Translat 2021; 30:41-50. [PMID: 34611513 PMCID: PMC8458725 DOI: 10.1016/j.jot.2021.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/03/2021] [Accepted: 08/08/2021] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE Synovial fibrosis is a characteristic symptom of osteoarthritis (OA), which is closely associated with joint pain and stiffness. Previous studies have reported that low-intensity pulsed ultrasound (LIPUS) can alleviate cartilage degradation in OA. However, the functions and mechanisms of LIPUS in OA synovial fibrosis are still unknown. METHODS The destabilization of the medial meniscus (DMM) mouse model of OA was established in C57 male mice and fibroblast-like synoviocytes (FLS) were isolated from synovial tissue of OA patients. The knee joint diameter, Masson's trichrome (MT) and Hematoxylin-eosin (HE) staining were used to evaluate synovial fibrosis and hyperplasia. The Immunohistochemistry (IHC) staining was performed to detected the expression of synovial fibrosis makers and the activation of Wnt/β-catenin signaling in vivo. FLS were treated with TGF-β1 to serve as an in vitro model of synovial fibrosis, Wnt3a was used to activate the Wnt/β-catenin signaling in cells. Cell proliferation was detected by using EdU assay, cell viability was performed by CCK8 assay. The protein levels of α-SMA, CTGF, Col Ⅰ, β-catenin, active β-catenin, c-Myc and cyclin D1 were examined by western blot and immunofluorescence staining. RESULTS Two weeks after the LIPUS treatment, the synovial fibrosis, synovial hyperplasia and synoviocyte proliferation in the DMM model were significantly decreased. In vitro, LIPUS directly inhibited the TGF-β1-induced fibrotic response and proliferation of FLS. Meanwhile, LIPUS suppressed Wnt/β-catenin signaling in the synovium of DMM mice and cultured FLS. More importantly, we found that the synovial fibrosis makers, Wnt/β-catenin pathway downstream proteins and FLS proliferation were significantly decreased in Wnt3a-stimulated FLS following LIPUS treatment. CONCLUSIONS Our results present a novel role of LIPUS in OA-related synovial fibrosis, which is associated with its ability to repress Wnt/β-catenin signaling in FLS. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE This study provides new insight into the clinical application of LIPUS as a therapeutic option to manage synovial fibrosis in OA.
Collapse
Affiliation(s)
- Bo Liao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Mengtong Guan
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
- Department of Rehabilitation Medicine, Chengdu Women's and Children's Central Hospital, Chengdu, Sichuan province, 610091, China
| | - Qiaoyan Tan
- Center of Bone Metabolism and Repair (Orthopedics Medicine), Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Gailan Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ruobin Zhang
- Center of Bone Metabolism and Repair (Orthopedics Medicine), Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Junlan Huang
- Center of Bone Metabolism and Repair (Orthopedics Medicine), Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Mi Liu
- Center of Bone Metabolism and Repair (Orthopedics Medicine), Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hong Chen
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Kaiting Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Dingqun Bai
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ying Zhu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
131
|
Ni J, Au M, Kong H, Wang X, Wen C. Lycium barbarum polysaccharides in ageing and its potential use for prevention and treatment of osteoarthritis: a systematic review. BMC Complement Med Ther 2021; 21:212. [PMID: 34404395 PMCID: PMC8371808 DOI: 10.1186/s12906-021-03385-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 07/29/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Lycium barbarum polysaccharide (LBP), the most abundant functional component of wolfberry, is considered a potent antioxidant and an anti-ageing substance. This review aims to outline the hallmarks of ageing in the pathogenesis of osteoarthritis (OA), followed by the current understanding of the senolytic effect of LBP and its potential use in the prevention and treatment of OA. This will be discussed through the lens of molecular biology and herbal medicine. METHODS A literature search was performed from inception to March 2020 using following keywords: "Lycium barbarum polysaccharide", "DNA damage", antioxidant, anti-apoptosis, anti-inflammation, anti-ageing, osteoarthritis, chondrocytes, fibroblasts, osteoblasts, osteoclasts, and "bone mesenchymal stem cell". The initial search yielded 2287 papers, from which 35 studies were selected for final analysis after screening for topic relevancy by the authors. RESULTS In literature different in vitro and in vivo ageing models are used to demonstrate LBP's ability to reduce oxidative stress, restore mitochondrial function, mitigate DNA damage, and prevent cellular senescence. All the evidence hints that LBP theoretically attenuates senescent cell accumulation and suppresses the senescence-associated secretory phenotype as observed by the reduction in pro-inflammatory cytokines, like interleukin-1beta, and matrix-degrading enzymes, such as MMP-1 and MMP-13. However, there remains a lack of evidence on the disease-modifying effect of LBP in OA, although its chondroprotective, osteoprotective and anti-inflammatory effects were reported. CONCLUSION Our findings strongly support further investigations into the senolytic effect of LBP in the context of age-related OA.
Collapse
Affiliation(s)
- Junguo Ni
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Manting Au
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Hangkin Kong
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Xinluan Wang
- Centre for Translational Medical Research and Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shen Zhen, China
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, Hong Kong Polytechnic University, Kowloon, Hong Kong.
| |
Collapse
|
132
|
You H, Zhang R, Wang L, Pan Q, Mao Z, Huang X. Chondro-Protective Effects of Shikimic Acid on Osteoarthritis via Restoring Impaired Autophagy and Suppressing the MAPK/NF-κB Signaling Pathway. Front Pharmacol 2021; 12:634822. [PMID: 34385915 PMCID: PMC8354550 DOI: 10.3389/fphar.2021.634822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a major cause of cartilage pain and limited mobility in middle-aged and elderly individuals. The degeneration of cartilage induced by inflammation and cartilage anabolic and catabolic disorder plays a key role in OA. Shikimic acid (SA), a natural ingredient extracted from Illicium verum, has been shown to exert notable anti-inflammatory effects in previous studies, suggesting its potential effects in the treatment of OA. In this study, we revealed that the pretreatment of SW1353 human chondrocytes with SA before interleukin 1β (IL-1β) stimulation effectively decreased the expression of inducible nitric oxide synthase (iNOS), cyclooxygenase (Cox)-2, matrix metalloproteinases (MMPs; MMP3 and MMP13), a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)-5, type X collagen, and p62; increased the expression of type II collagen, ATG7, Beclin-1, and LC3; and increased the autophagic flux. Mechanistically, we found that SA suppressed the IL-1β-induced activation of the mitogen-activated protein kinase (MAPK) and nuclear factor-kappaB (NF-κB) pathways. Furthermore, the results of safranin O staining and toluidine blue staining of primary rat cartilage chondrocytes and a trauma-induced rat model of OA showed that SA alleviated progression of OA in vivo. Collectively, our research enhances understanding of the mechanism of protective effect of SA against the progression of OA, which involves amelioration of cartilage degeneration, thereby providing new evidence for the use of SA as a therapy to prevent the development of OA.
Collapse
Affiliation(s)
- Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingying Wang
- Department of Radiation Oncology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiyong Pan
- Department of Orthopaedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zekai Mao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojian Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
133
|
Kim D, Song J, Jin EJ. BNIP3-Dependent Mitophagy via PGC1α Promotes Cartilage Degradation. Cells 2021; 10:cells10071839. [PMID: 34360007 PMCID: PMC8304751 DOI: 10.3390/cells10071839] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/14/2022] Open
Abstract
Since mitochondria are suggested to be important regulators in maintaining cartilage homeostasis, turnover of mitochondria through mitochondrial biogenesis and mitochondrial degradation may play an important role in the pathogenesis of osteoarthritis (OA). Here, we found that mitochondrial dysfunction is closely associated with OA pathogenesis and identified the peroxisome proliferator-activated receptor-gamma co-activator 1-alpha (PGC1α) as a potent regulator. The expression level of PGC1α was significantly decreased under OA conditions, and knockdown of PGC1α dramatically elevated the cartilage degradation by upregulating cartilage degrading enzymes and apoptotic cell death. Interestingly, the knockdown of PGC1α activated the parkin RBR E3 ubiquitin protein ligase (PRKN)-independent selective mitochondria autophagy (mitophagy) pathway through the upregulation of BCL2 and adenovirus E1B 19-kDa-interacting protein 3 (BNIP3). The overexpression of BNIP3 stimulated mitophagy and cartilage degradation by upregulating cartilage-degrading enzymes and chondrocyte death. We identified microRNA (miR)-126-5p as an upstream regulator for PGC1α and confirmed the direct binding between miR-126-5p and 3′ untranslated region (UTR) of PGC1α. An in vivo OA mouse model induced by the destabilization of medial meniscus (DMM) surgery, and the delivery of antago-miR-126 via intra-articular injection significantly decreased cartilage degradation. In sum, the loss of PGC1α in chondrocytes due to upregulation of miR-126-5p during OA pathogenesis resulted in the activation of PRKN-independent mitophagy through the upregulation of BNIP3 and stimulated cartilage degradation and apoptotic death of chondrocytes. Therefore, the regulation of PGC1α:BNIP3 mitophagy axis could be of therapeutic benefit to cartilage-degrading diseases.
Collapse
MESH Headings
- Animals
- Antagomirs/genetics
- Antagomirs/metabolism
- Arthroplasty, Replacement, Knee/methods
- Base Sequence
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Menisci, Tibial/metabolism
- Menisci, Tibial/pathology
- Mice
- Mice, Inbred C57BL
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/metabolism
- Mitophagy/genetics
- Osteoarthritis/genetics
- Osteoarthritis/metabolism
- Osteoarthritis/pathology
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Primary Cell Culture
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Signal Transduction
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Deokha Kim
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (D.K.); (J.S.)
| | - Jinsoo Song
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (D.K.); (J.S.)
- Integrated Omics Institute, Wonkwang University, Iksan 54538, Jeonbuk, Korea
| | - Eun-Jung Jin
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan 54538, Jeonbuk, Korea; (D.K.); (J.S.)
- Integrated Omics Institute, Wonkwang University, Iksan 54538, Jeonbuk, Korea
- Correspondence: ; Tel.: +82-63-850-6192; Fax: +82-63-850-6197
| |
Collapse
|
134
|
Zhou W, Shi Y, Wang H, Yu C, Zhu H, Wu A. Sinensetin Reduces Osteoarthritis Pathology in the Tert-Butyl Hydroperoxide-Treated Chondrocytes and the Destabilization of the Medial Meniscus Model Mice via the AMPK/mTOR Signaling Pathway. Front Pharmacol 2021; 12:713491. [PMID: 34335275 PMCID: PMC8322586 DOI: 10.3389/fphar.2021.713491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/06/2021] [Indexed: 12/25/2022] Open
Abstract
As a common degenerative disease, osteoarthritis (OA) usually causes disability in the elderly and socioeconomic burden. Previous studies have shown that proper autophagy has a protective effect on OA. Sinensetin (Sin) is a methylated flavonoid derived from citrus fruits. Studies have shown that Sin is a good autophagy inducer and has shown excellent therapeutic effects in a variety of diseases; however, its role in the treatment of OA is not fully understood. This study proved the protective effect of Sin on OA through a series of in vivo and in vitro experiments. In vitro experiments have shown that Sin may inhibit chondrocyte apoptosis induced by tert-butyl hydroperoxide (TBHP); at the same time, it might also inhibit the production of MMP13 and promote the production of aggrecan and collagen II. Mechanism studies have shown that Sin promotes chondrocyte autophagy by activating AMPK/mTOR signaling pathway. On the contrary, inhibition of autophagy can partially abolish the protective effect of Sin on TBHP-treated chondrocytes. In vivo experiments show that Sin may protect against DMM-induced OA pathogenesis. These results provide evidence that Sin serves as a potential candidate for the treatment of OA.
Collapse
Affiliation(s)
- Wenxian Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Caiyu Yu
- The School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Huanqing Zhu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
135
|
Enzymatic Machinery of Ubiquitin and Ubiquitin-Like Modification Systems in Chondrocyte Homeostasis and Osteoarthritis. Curr Rheumatol Rep 2021; 23:62. [PMID: 34216299 DOI: 10.1007/s11926-021-01022-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW To date, a vast amount of information regarding ubiquitination (Ub) and ubiquitylation-like (Ubl) modification-related mechanisms has been reported in the context of skeletal cell homeostasis and diseases. In this review, we mainly focus on recent findings regarding the contribution of enzymatic machinery that directly adds or removes Ub and Ubl modifications from protein targets in chondrocyte homeostasis and osteoarthritis (OA) development. RECENT FINDINGS Mechanisms that promote homeostasis of articular chondrocytes are crucial for maintaining the integrity of articular joints to prevent osteoarthritis development. Articular chondrocytes are postmitotic cells that continuously produce and remodel cartilage matrix. In addition, the long lifespan of chondrocytes makes them susceptible to accumulating cellular damage. Ub and the evolutionarily conserved Ubl modifications, such as SUMOylation, ATGylation, and UFMylation, play important roles in promoting chondrocyte homeostasis, including regulating cell signaling and protein stability, resolving cellular stresses and inflammation, and maintaining differentiation and survival of chondrocytes. Uncovering new components/functions of Ub/Ubl modification machinery may provide novel drug targets to treat OA.
Collapse
|
136
|
Yi D, Yu H, Lu K, Ruan C, Ding C, Tong L, Zhao X, Chen D. AMPK Signaling in Energy Control, Cartilage Biology, and Osteoarthritis. Front Cell Dev Biol 2021; 9:696602. [PMID: 34239878 PMCID: PMC8258395 DOI: 10.3389/fcell.2021.696602] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022] Open
Abstract
The adenosine monophosphate (AMP)-activated protein kinase (AMPK) was initially identified as an enzyme acting as an "energy sensor" in maintaining energy homeostasis via serine/threonine phosphorylation when low cellular adenosine triphosphate (ATP) level was sensed. AMPK participates in catabolic and anabolic processes at the molecular and cellular levels and is involved in appetite-regulating circuit in the hypothalamus. AMPK signaling also modulates energy metabolism in organs such as adipose tissue, brain, muscle, and heart, which are highly dependent on energy consumption via adjusting the AMP/ADP:ATP ratio. In clinics, biguanides and thiazolidinediones are prescribed to patients with metabolic disorders through activating AMPK signaling and inhibiting complex I in the mitochondria, leading to a reduction in mitochondrial respiration and elevated ATP production. The role of AMPK in mediating skeletal development and related diseases remains obscure. In this review, in addition to discuss the emerging advances of AMPK studies in energy control, we will also illustrate current discoveries of AMPK in chondrocyte homeostasis, osteoarthritis (OA) development, and the signaling interaction of AMPK with other pathways, such as mTOR (mechanistic target of rapamycin), Wnt, and NF-κB (nuclear factor κB) under OA condition.
Collapse
Affiliation(s)
- Dan Yi
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huan Yu
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ke Lu
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changshun Ruan
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Liping Tong
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiaoli Zhao
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Di Chen
- Faculty of Pharmaceutical Sciences, Shenzhen, China
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
137
|
|
138
|
Maglaviceanu A, Wu B, Kapoor M. Fibroblast-like synoviocytes: Role in synovial fibrosis associated with osteoarthritis. Wound Repair Regen 2021; 29:642-649. [PMID: 34021514 DOI: 10.1111/wrr.12939] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/02/2021] [Accepted: 05/08/2021] [Indexed: 12/12/2022]
Abstract
The synovial membrane undergoes a variety of structural changes throughout the pathogenesis of osteoarthritis (OA), including the development of fibrosis. Fibroblast-like synoviocytes (FLS) are a heterogenous cell population of the synovium that are suggested to drive the fibrotic response, but the exact mechanisms associated with their activation in OA remain unclear. Once activated, FLS are suggested to acquire a myofibroblast-like phenotype that drives fibrogenesis through excessive extracellular matrix (ECM) component deposition and an enhanced contractile function. In this review, we define FLS in the synovium, discuss how select extracellular or endogenous factors potentially induce their activation in OA, and describe how the activity of myofibroblast-like cells affects the structure of the synovial membrane.
Collapse
Affiliation(s)
- Anca Maglaviceanu
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Brian Wu
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
139
|
Wang S, Mobasheri A, Zhang Y, Wang Y, Dai T, Zhang Z. Exogenous stromal cell-derived factor-1 (SDF-1) suppresses the NLRP3 inflammasome and inhibits pyroptosis in synoviocytes from osteoarthritic joints via activation of the AMPK signaling pathway. Inflammopharmacology 2021; 29:695-704. [PMID: 34085175 PMCID: PMC8233244 DOI: 10.1007/s10787-021-00814-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/24/2021] [Indexed: 01/21/2023]
Abstract
Objective NLRP3 inflammasome may play a key role in OA pathogenesis. Stromal cell-derived factor-1 (SDF-1) is a homeostatic CXC chemokine. Since the role of SDF-1 in OA has not been explored, this study aimed to examine the effect of SDF-1 on NLRP3 inflammasome and pyroptosis in synoviocytes from OA joints. Materials and methods Human synovium was obtained from OA patients for isolation of primary synoviocytes and a murine model of collagenase-induced OA was established for testing intra-articular injections of SDF-1. Immunoblotting assays were used to examine the effects and underlying mechanism of action of SDF-1 on NLRP3 inflammasome and synoviocyte pyroptosis in synoviocytes. Inhibitors of AMPK and PI3K–mTOR were utilized to investigate the key signaling pathways involved in SDF-1-mediated OA inflammasome formation and pyroptosis. Results Synoviocytes from OA joints exhibited significantly higher expression of NLRP3 inflammasome and biomarkers of synoviocyte pyroptosis relative to healthy individuals. This was confirmed in the collagenase-induced OA model, where OA synoviocytes had a significantly lower SDF-1 expression than healthy ones. SDF-1 treatment in synoviocytes of OA patients and collagenase-induced OA led to significant downregulation in the expression of NLRP3 inflammasome and synoviocyte pyroptosis biomarkers. Inhibition of the AMPK signaling pathway significantly suppressed the inhibitory effect of SDF-1 on NLRP3 inflammasome expression of OA synoviocytes. However, blocking the SDF-1-activated PI3K–mTOR signaling pathway could still suppress the expression of NLRP3 inflammasome and synoviocyte pyroptosis biomarkers. Conclusions SDF-1 ameliorates NLRP3 inflammasome and pyroptosis in OA synoviocytes through activation of the AMPK signaling pathway. Therefore, SDF-1 may be a novel therapeutic target for OA. Supplementary Information The online version contains supplementary material available at 10.1007/s10787-021-00814-x.
Collapse
Affiliation(s)
- Shuya Wang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng St, Harbin, 150001, China
| | - Ali Mobasheri
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China. .,Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, PO Box 5000, 90014, Oulu, Finland. .,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania. .,Department of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, 508 GA, Utrecht, The Netherlands.
| | - Yue Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng St, Harbin, 150001, China
| | - Yanli Wang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng St, Harbin, 150001, China
| | - Tianqi Dai
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng St, Harbin, 150001, China
| | - Zhiyi Zhang
- Department of Rheumatology, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng St, Harbin, 150001, China.
| |
Collapse
|
140
|
Tao Y, Zhou J, Wang Z, Tao H, Bai J, Ge G, Li W, Zhang W, Hao Y, Yang X, Geng D. Human bone mesenchymal stem cells-derived exosomal miRNA-361-5p alleviates osteoarthritis by downregulating DDX20 and inactivating the NF-κB signaling pathway. Bioorg Chem 2021; 113:104978. [PMID: 34052737 DOI: 10.1016/j.bioorg.2021.104978] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/05/2021] [Accepted: 05/04/2021] [Indexed: 12/18/2022]
Abstract
Osteoarthritis (OA) is a chronic disease featured by joint hyperplasia, deterioration of articular cartilage, and progressive degeneration. Abnormal expression of microRNAs (miRNAs) has been found to be implicated in the pathological process of OA. In this study, the role of miR-361-5p transferred by exosomes derived from human bone mesenchymal stem cells (hBMSCs) in OA was investigated. The expression of Asp-Glu-Ala-Asp-box polypeptide 20 (DDX20) and miR-361-5p in interleukin-1β (IL-1β)-treated chondrocytes was determined by reverse transcription quantitative polymerase chain reaction. DDX20 was knocked down by transfection of short hairpin RNA targeting DDX20, and the effects of DDX20 downregulation on IL-1β-induced damage of chondrocytes were detected. The interaction between DDX20 and miR-361-5p was tested by luciferase report assay. hBMSCs-derived exosomes loaded with miR-361-5p were co-incubated with chondrocytes followed by detection of cell viability, proliferation and inflammatory response. An OA rat model was established to further explore the role of miR-361-5p in vivo. Western blot, luciferase reporter and immunofluorescence staining assays were used to evaluate the activation of the nuclear factor kappa-B (NF-κB) signaling pathway. We found that DDX20 was upregulated, while miR-361-5p was underexpressed in IL-1β-treated chondrocytes. Downregulation of DDX20 inhibits levels of matrix metalloproteinases (MMPs) and suppresses inflammation induced by IL-1β. Mechanistically, miR-361-5p was verified to directly target DDX20. In addition, hBMSC-derived exosomes-transferred miR-361-5p alleviates chondrocyte damage and inhibits the NF-κB signaling pathway via targeting DDX20. Inhibition of NF-κB signaling reverses the effect of overexpressed DDX20 on IL-1β-induced chondrocyte damage. Moreover, exosomal miR-361-5p alleviates OA damage in vivo. Overall, hBMSC-derived exosomal miR-361-5p alleviates OA damage by targeting DDX20 and inactivating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yunxia Tao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215006, Jiangsu, China
| | - Zhen Wang
- Department of Orthopaedics, Suzhou Kowloon Hospital Shanghai Jiao Tong University School of Medicine, Suzhou 215006, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Wei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215006, Jiangsu, China.
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215006, Jiangsu, China.
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
141
|
Perez-Tejeiro JM, Csukasi F. DEPTOR in Skeletal Development and Diseases. Front Genet 2021; 12:667283. [PMID: 34122519 PMCID: PMC8191632 DOI: 10.3389/fgene.2021.667283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/12/2021] [Indexed: 12/02/2022] Open
Abstract
Discovered in 2009, the DEP-domain containing mTOR-interacting protein, DEPTOR, is a known regulator of the mechanistic target of rapamycin (mTOR), an evolutionarily conserved kinase that regulates diverse cellular processes in response to environmental stimuli. DEPTOR was originally identified as a negative regulator of mTOR complexes 1 (mTORC1) and 2 (mTORC2). However, recent discoveries have started to unravel the roles of DEPTOR in mTOR-independent responses. In the past few years, mTOR emerged as an important regulator of skeletal development, growth, and homeostasis; the dysregulation of its activity contributes to the development of several skeletal diseases, both chronic and genetic. Even more recently, several groups have reported on the relevance of DEPTOR in skeletal biology through its action on mTOR-dependent and mTOR-independent pathways. In this review, we summarize the current understanding of DEPTOR in skeletal development and disease.
Collapse
Affiliation(s)
- Jose Miguel Perez-Tejeiro
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, IBIMA, University of Málaga, Málaga, Spain.,Biomaterials and Nanomedicine (CIBER-BBN), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, Networking Biomedical Research Center in Bioengineering, Málaga, Spain
| | - Fabiana Csukasi
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, IBIMA, University of Málaga, Málaga, Spain.,Biomaterials and Nanomedicine (CIBER-BBN), Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, Networking Biomedical Research Center in Bioengineering, Málaga, Spain
| |
Collapse
|
142
|
Stimulation of α7-nAChRs coordinates autophagy and apoptosis signaling in experimental knee osteoarthritis. Cell Death Dis 2021; 12:448. [PMID: 33953172 PMCID: PMC8100296 DOI: 10.1038/s41419-021-03726-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/30/2022]
Abstract
Osteoarthritis (OA) is the most common chronic joint disease in the elderly population. Growing evidence indicates that a balance between autophagy and apoptosis in chondrocytes plays a key role in OA’s cartilage degradation. Thus, drugs targeting the balance between apoptosis and autophagy are potential therapeutic approaches for OA treatment. In previous studies, we found that the activation of α7 nicotinic acetylcholine receptors (α7-nAChRs) alleviated monosodium iodoacetate (MIA)-induced joint degradation and osteoarthritis pain. To explore the potential functions of α7-nAChRs in autophagy and apoptosis signaling in knee OA, we compared the expression of α7-nAChRs in human knee articular cartilage tissues from normal humans and OA patients. We found that knee joint cartilage tissues of OA patients showed decreased α7-nAChRs and an imbalance between autophagy and apoptosis. Next, we observed that α7-nAChRs deficiency did not affect cartilage degradation in OA development but reversed the beneficial effects of nicotine on mechanical allodynia, cartilage degradation, and an MIA-induced switch from autophagy to apoptosis. Unlike in vivo studies, we found that primary chondrocytes from α7-nAChRs knockout (KO) mice showed decreased LC3 levels under normal conditions and were more sensitive toward MIA-induced apoptosis. Finally, we found that α7-nAChRs deficiency increased the phosphorylation of mTOR after MIA treatment, which can also be observed in OA patients’ tissues. Thus, our findings not only confirmed that nicotine alleviated MIA-induced pain behavior and cartilage degradation via stimulating the α7-nAChRs/mTOR signal pathway but found the potential role of α7-nAChRs in mediating the balance between apoptosis and autophagy.
Collapse
|
143
|
Liu H, Zhu H, Cheng L, Zhao Y, Chen X, Li J, Xv X, Xiao Z, Li W, Pan J, Zhang Q, Zeng C, Guo J, Xie D, Cai D. TCP/PLGA composite scaffold loaded rapamycin in situ enhances lumbar fusion by regulating osteoblast and osteoclast activity. J Tissue Eng Regen Med 2021; 15:475-486. [PMID: 33686790 DOI: 10.1002/term.3186] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/22/2021] [Indexed: 11/07/2022]
Abstract
The purpose of this study was to develop a novel β-tricalcium phosphate (TCP)/poly (D,L-lactic-co-glycolic acid) (PLGA) composite scaffold loaded with rapamycin that can regulate the activity of osteoblasts and osteoclasts for lumbar fusion. The TCP/PLGA composite scaffold was fabricated by cryogenic three-dimensional printing techniques and then loaded with rapamycin in situ. The structural surface morphology of the composite scaffold was tested with scanning electron microscope. To evaluate the biocompatibility of the composite scaffold in vitro, bone marrow mesenchymal stem cells (BMSCs) were cultured on the TCP/PLGA composite scaffold slide and tested with Live/Dead Viability Kit. The effect of rapamycin on osteoclast and osteoblast was studied with staining and Western blotting. The in vitro results showed that the rapamycin-loaded TCP/PLGA composite scaffold showed good biocompatibility with BMSC and released rapamycin obviously promoted the osteoblast differentiation and mineralization. In vivo study, the TCP/PLGA composite scaffold loaded with rapamycin were implanted in lumbar fusion model and study with micro-computed tomography scanning, hematoxylin-eosin, Masson, and immune-histological staining, to evaluate the effect of rapamycin on bone fusion. The in vivo results demonstrated that rapamycin-loaded TCP/PLGA composite scaffold could enhance bone formation by regulating osteoblast and osteoclast activity, respectively. In this study, the TCP/PLGA composite scaffold loaded with rapamycin was confirmed to provide great compatibility and improved performance in lumbar fusion by regulating osteoblastic and osteoclastic activity and would be a promising composite biomaterial for bone tissue engineering.
Collapse
Affiliation(s)
- Hai Liu
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Huangrong Zhu
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Liang Cheng
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yitao Zhao
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xizhong Chen
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jintao Li
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Xv
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhisheng Xiao
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Li
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jianying Pan
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Qun Zhang
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Office of Clinical Trial of Drug, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Chun Zeng
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jinshan Guo
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Histology and Embryology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, China
| | - Denghui Xie
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Daozhang Cai
- Department of Orthopaedic Surgery, Center for Orthopaedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
144
|
Li Z, Wang J, Deng X, Huang D, Shao Z, Ma K. Compression stress induces nucleus pulposus cell autophagy by inhibition of the PI3K/AKT/mTOR pathway and activation of the JNK pathway. Connect Tissue Res 2021; 62:337-349. [PMID: 32180463 DOI: 10.1080/03008207.2020.1736578] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: Reactive oxygen species (ROS) are related to compression stress-induced nucleus pulposus (NP) cell autophagy, but the specific mechanism is unknown in compression stress-induced intervertebral disc degeneration (IVDD). Here, we discuss the specific molecular mechanism and explore whether ROS scavengers could be employed as specific drugs to inhibit compression stress-induced IVDD.Methods: Rat NP cells were exposed to 1.0 MPa compression and pretreatment with the ROS scavenger N-acetylcysteine (NAC) or the JNK-selective inhibitor SP600125 not. Intracellular ROS production was monitored by confocal microscopy. Autophagy was detected by observing the NP cell ultrastructural features using TEM and examining autophagic vacuoles by flow cytometry. The levels of autophagy-associated molecules, the JNK pathway and the PI3K/AKT/mTOR pathway were analyzed by western blotting.Results: Compression-mediated autophagy in rat NP cells was implicated in ROS generation. The ROS scavenger NAC could protect compression-induced NP cell injures by inhibiting ROS production. And SP600125, a JNK inhibitor, attenuated compression-induced NP cell autophagy. Additionally, this is the first report showing that compression induces autophagy in rat NP cells by impeding the compression-induced ROS dependent PI3K/AKT/mTOR pathway and the ROS independent activation of JNK pathway. And the involvement of JNK pathway was in different mechanism of action that when inhibited leaded to increased cell death, increased generation of ROS but decreased autophagy.Conclusions: These results show a new regulatory mechanism involving ROS-mediated autophagy in rat NP cells, which may provide ideas for drug development to improve compression stress-induced IVDD and help avoid eventual surgical treatment of IVD herniation.
Collapse
Affiliation(s)
- Zhiliang Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Wang
- Department of Gastroenterology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyu Deng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donghua Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaige Ma
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
145
|
Tripolino C, Ciaffi J, Pucino V, Ruscitti P, van Leeuwen N, Borghi C, Giacomelli R, Meliconi R, Ursini F. Insulin Signaling in Arthritis. Front Immunol 2021; 12:672519. [PMID: 33995414 PMCID: PMC8119635 DOI: 10.3389/fimmu.2021.672519] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/12/2021] [Indexed: 12/23/2022] Open
Abstract
Inflammatory arthritis is burdened by an increased risk of metabolic disorders. Cytokines and other mediators in inflammatory diseases lead to insulin resistance, diabetes and hyperlipidemia. Accumulating evidence in the field of immunometabolism suggests that the cause-effect relationship between arthritis and metabolic abnormalities might be bidirectional. Indeed, the immune response can be modulated by various factors such as environmental agents, bacterial products and hormones. Insulin is produced by pancreatic cells and regulates glucose, fat metabolism and cell growth. The action of insulin is mediated through the insulin receptor (IR), localized on the cellular membrane of hepatocytes, myocytes and adipocytes but also on the surface of T cells, macrophages, and dendritic cells. In murine models, the absence of IR in T-cells coincided with reduced cytokine production, proliferation, and migration. In macrophages, defective insulin signaling resulted in enhanced glycolysis affecting the responses to pathogens. In this review, we focalize on the bidirectional cause-effect relationship between impaired insulin signaling and arthritis analyzing how insulin signaling may be involved in the aberrant immune response implicated in arthritis and how inflammatory mediators affect insulin signaling. Finally, the effect of glucose-lowering agents on arthritis was summarized.
Collapse
Affiliation(s)
- Cesare Tripolino
- Geriatric Medicine Unit, Department of Medical Functional Area, "San Giovanni di Dio" Hospital, Crotone, Italy
| | - Jacopo Ciaffi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli (IOR), Bologna, Italy
| | - Valentina Pucino
- Institute of Inflammation and Ageing, University of Birmingham and Queen Elizabeth Hospital, Birmingham, United Kingdom
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nina van Leeuwen
- Rheumatology Department, Leiden University Medical Center, Leiden, Netherlands
| | - Claudio Borghi
- Unità Operativa Medicina Interna Cardiovascolare-IRCCS Azienda Ospedaliera-Universitaria, Bologna, Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome "Campus Biomedico", Rome, Italy
| | - Riccardo Meliconi
- Geriatric Medicine Unit, Department of Medical Functional Area, "San Giovanni di Dio" Hospital, Crotone, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Francesco Ursini
- Geriatric Medicine Unit, Department of Medical Functional Area, "San Giovanni di Dio" Hospital, Crotone, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
146
|
Sun L, Zheng W, Liu QD, Ge L. Valproic Acid Protects Chondrocytes from LPS-Stimulated Damage via Regulating miR-302d-3p/ITGB4 Axis and Mediating the PI3K-AKT Signaling Pathway. Front Mol Biosci 2021; 8:633315. [PMID: 33968981 PMCID: PMC8100442 DOI: 10.3389/fmolb.2021.633315] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/04/2021] [Indexed: 01/10/2023] Open
Abstract
Background: Osteoarthritis (OA) is one of the most common degenerative joint diseases characterized by increased apoptosis and autophagy deficiency. The investigation was performed to examine the effect of valproic acid (VPA) and molecular mechanism related to miR-302d-3p/ITGB4 axis in OA. Methods: The OA clinical samples were obtained from the GEO database to analyze differentially expressed genes. An in vitro OA model was mimicked by LPS in CHON-001 cells. Autophagy-related genes were downloaded from the HADb website, and potential drugs were mined using the CTD website. The upstream factors of ITGB4 were predicted with bioinformatics analysis, which was validated by luciferase activity assay and RIP assay. Cell viability and apoptosis were evaluated using CCK-8 and flow cytometry. The expression levels, including ITGB4, miR-302d-3p, and autophagy-/PI3K-AKT pathway-related markers, were measured by qRT-PCR or/and western blot. Results: Our results showed that miR-302d-3p inhibited cell viability and promoted apoptosis of LPS-treated CHON-001 cells by targeting ITGB4. VPA treatment remarkably alleviated LPS-stimulated injury in CHON-001 cells. The inhibitory effect of VPA on LPS-stimulated damage in CHON-001 cells was weakened by miR-302d-3p overexpression, while it was intensified because of ITGB4 upregulation. Mechanistically, VPA treatment induced a significant decrease in the levels of p-PI3K and p-AKT in LPS-stimulated CHON-001 cells through regulating miR-302d-3p/ITGB4 axis. Conclusion: Overall, VPA treatment may ameliorate LPS-induced injury on chondrocytes via the regulation of miR-302d-3p/ITGB4 pair and the inactivation of the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Long Sun
- Department of Orthopedics, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, China
| | - Wei Zheng
- Department of Joint Surgery, Rizhao Central Hospital, Rizhao, China
| | - Qian-Dong Liu
- Department of Emergency, People's Hospital of Rizhao, Rizhao, China
| | - Lei Ge
- Department of Emergency, People's Hospital of Rizhao, Rizhao, China
| |
Collapse
|
147
|
Dai S, Wang H, Wang M, Zhang Y, Zhang Z, Lin Z. Comparative transcriptomics and network pharmacology analysis to identify the potential mechanism of celastrol against osteoarthritis. Clin Rheumatol 2021; 40:4259-4268. [PMID: 33870466 DOI: 10.1007/s10067-021-05726-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Celastrol is a promising therapeutic agent for the treatment of osteoarthritis (OA). However, the mechanism of action of celastrol is unclear. This study was aiming to identify the potential function of celastrol on OA and determine its underlying mechanism. METHOD Celastrol targets were collected from web database searches and literature review, while pathogenic OA targets were obtained from Online Mendelian Inheritance in Man (OMIM) and GeneCards databases. Transcriptomics data was sequenced using an Illumina HiSeq 4000 platform. Celastrol-OA overlapping genes were then identified followed by prediction of the potential function and signaling pathways associated with celastrol using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. A celastrol-target network was constructed to identify the candidate core targets of celastrol. The predictions were then validated by performing molecular docking and molecular dynamics simulation studies. RESULTS In total, 96 genes were identified as the putative celastrol targets for treatment of OA. These genes were possibly involved in cell phenotype changes including response to lipopolysaccharide and oxidative stress as well as in cell apoptosis and aging. The genes also induced the mTOR pathway and AGE-RAGE signaling pathway at the intracellular level. Additionally, results indicated that 13 core targets including mTOR, TP53, MMP9, EGFR, CCND1, MAPK1, STAT3, VEGFA, CASP3, TNF, MYC, ESR1, and PTEN were likely direct targets of celastrol in OA. Finally, mTOR was determined as the most likely therapeutic target of celastrol in OA. CONCLUSION This study provides a basic understanding and novel insight into the potential mechanism of celastrol against OA. Key Points • Our study provides a strong indication that further study of celastrol therapy in OA is required. • mTOR is the most likely therapeutic target of celastrol in OA.
Collapse
Affiliation(s)
- Siming Dai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Hui Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Meng Wang
- Key Laboratory of Basic and Applied Research in North Medicine, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yue Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Zhiyi Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.
| | - Zhiguo Lin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang, China.
| |
Collapse
|
148
|
Inhibition of Autophagy at Different Stages by ATG5 Knockdown and Chloroquine Supplementation Enhances Consistent Human Disc Cellular Apoptosis and Senescence Induction rather than Extracellular Matrix Catabolism. Int J Mol Sci 2021; 22:ijms22083965. [PMID: 33921398 PMCID: PMC8069032 DOI: 10.3390/ijms22083965] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/30/2021] [Accepted: 04/08/2021] [Indexed: 12/27/2022] Open
Abstract
The intervertebral disc is the largest avascular organ. Autophagy is an important cell survival mechanism by self-digestion and recycling damaged components under stress, primarily nutrient deprivation. Resident cells would utilize autophagy to cope with the harsh disc environment. Our objective was to elucidate the roles of human disc cellular autophagy. In human disc cells, serum deprivation and pro-inflammatory interleukin-1β (IL-1β) stimulation increased autophagy marker microtubule-associated protein 1 light chain 3 (LC3)-II and decreased autophagy substrate p62/sequestosome 1 (p62/SQSTM1), indicating enhanced autophagy. Then, RNA interference (RNAi) of autophagy-related gene 5 (ATG5), essential for autophagy, showed decreases in ATG5 protein (26.8%–27.4%, p < 0.0001), which suppressed early-stage autophagy with decreased LC3-II and increased p62/SQSTM1. Cell viability was maintained by ATG5 RNAi in serum-supplemented media (95.5%, p = 0.28) but reduced in serum-free media (80.4%, p = 0.0013) with IL-1β (69.9%, p = 0.0008). Moreover, ATG5 RNAi accelerated IL-1β-induced changes in apoptosis and senescence. Meanwhile, ATG5 RNAi unaffected IL-1β-induced catabolic matrix metalloproteinase release, down-regulated anabolic gene expression, and mitogen-activated protein kinase pathway activation. Lysosomotropic chloroquine supplementation presented late-stage autophagy inhibition with apoptosis and senescence induction, while catabolic enzyme production was modest. Disc-tissue analysis detected age-related changes in ATG5, LC3-II, and p62/SQSTM1. In summary, autophagy protects against human disc cellular apoptosis and senescence rather than extracellular matrix catabolism.
Collapse
|
149
|
Zhou H, Li G, Wang Y, Jiang R, Li Y, Wang H, Wang F, Ma H, Cao L. Microbial Metabolite Sodium Butyrate Attenuates Cartilage Degradation by Restoring Impaired Autophagy and Autophagic Flux in Osteoarthritis Development. Front Pharmacol 2021; 12:659597. [PMID: 33897442 PMCID: PMC8062861 DOI: 10.3389/fphar.2021.659597] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease with multiple etiologies that affects individuals worldwide. No effective interventions are currently available to reverse the pathological process of OA. Sodium butyrate (NaB), a component of short-chain fatty acids (SCFAs), has multiple biological activities, including the attenuation of inflammation and anti-tumor activities in various diseases. However, whether the protective effects of NaB in OA are associated with the promotion of autophagy had not been investigated. Here, we explored the chondroprotective properties of NaB in an interleukin (IL)-1β-induced inflammatory chondrocyte model and an anterior cruciate ligament transection (ACLT) mouse model. Hematoxylin and eosin (HE), Safranin O, and immunohistochemical staining were performed to evaluate the effects of NaB treatment on articular cartilage. An optimal NaB dose for chondrocyte treatment was determined via cell counting kit-8 assays. Immunofluorescence and transmission electron microscopy were used to detect autophagy in chondrocytes. Flow cytometry was utilized to detect reactive oxygen species (ROS), cell cycle activity, and apoptosis in chondrocytes. Western blot and immunostaining were performed to evaluate the protein expression levels of relevant indicators. We found that the administration of NaB by oral gavage could attenuate cartilage degradation. In parallel, NaB treatment could enhance the activation of autophagy, increase autophagic flux, decrease extracellular matrix degradation, and reduce apoptosis by restraining inflammation, ROS production, and cell cycle arrest in IL-1β-treated chondrocytes. The protective effects of NaB could be partially abolished by the autophagy inhibitor 3-methyladenine (3-MA), which indicated that the protective effects of NaB against OA were partially governed by the enhancement of autophagy to restrain the formation of inflammatory mediators and ROS and regulate cell cycle progression and apoptosis in chondrocytes. In conclusion, NaB could attenuate OA progression by restoring impaired autophagy and autophagic flux via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway, both in vitro and in vivo, implying that NaB could represent a novel therapeutic approach for OA.
Collapse
Affiliation(s)
- Haikang Zhou
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Guoqing Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yang Wang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Rendong Jiang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yicheng Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Huhu Wang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fei Wang
- Xinjiang Uygur Autonomous Region Clinical Research Center for Orthopedic Diseases, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hairong Ma
- Xinjiang Uygur Autonomous Region Clinical Research Center for Orthopedic Diseases, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
150
|
Down-regulation of protease-activated receptor 2 ameliorated osteoarthritis in rats through regulation of MAPK/NF-κB signaling pathway in vivo and in vitro. Biosci Rep 2021; 40:222298. [PMID: 32134473 PMCID: PMC7098131 DOI: 10.1042/bsr20192620] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 01/09/2023] Open
Abstract
Recently, protease-activated receptor 2 (PAR2) has been proved to be involved in the inflammatory response including osteoarthritis (OA). In the present study, we found that PAR2 antagonist could remarkably improve the pathological condition of OA rats in vivo. In addition, we also found that PAR2 antagonist could suppress the production of inflammatory factors (TNF-α and Cox-2), decrease the levels of MMP-1 and MMP-13, and restrain the levels of P62 proteins and aggravate the expression of LC3-II both in vivo and in vitro. Besides, in vitro, PAR2 antagonist could increase the proliferation and colony formation of chondrocytes induced with IL-1β. Moreover, PAR2 antagonist could decrease the expression of expressions of p-p38, p-IκBα and p-NF-κB in vitro. However, PAR2 agonist exhibited the opposite effects. Furthermore, SB203580, a p38 MAPK inhibitor, could remarkably promote the proliferation of chondrocytes induced with IL-1β, could alleviate the production of TNF-α and Cox-2, could down-regulate the protein expressions of MMP-1 and MMP-13, and could decrease the expression of P62 and increase the expressions of LC3-II of chondrocytes induced with IL-1β. Importantly, SB203580 could reverse the effects of PAR2 agonist on the functions of chondrocytes induced with IL-1β. Taken together, the present data suggest that down-regulation of PAR2 can ameliorate OA through inducing autophagy via regulation of MAPK/NF-κB signaling pathway in vivo and in vitro, and PAR2 can be considered as a potential candidate to treat OA.
Collapse
|