51
|
Takahata Y, Hagino H, Kimura A, Urushizaki M, Yamamoto S, Wakamori K, Murakami T, Hata K, Nishimura R. Regulatory Mechanisms of Prg4 and Gdf5 Expression in Articular Cartilage and Functions in Osteoarthritis. Int J Mol Sci 2022; 23:ijms23094672. [PMID: 35563063 PMCID: PMC9105027 DOI: 10.3390/ijms23094672] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Owing to the rapid aging of society, the numbers of patients with joint disease continue to increase. Accordingly, a large number of patients require appropriate treatment for osteoarthritis (OA), the most frequent bone and joint disease. Thought to be caused by the degeneration and destruction of articular cartilage following persistent and excessive mechanical stimulation of the joints, OA can significantly impair patient quality of life with symptoms such as knee pain, lower limb muscle weakness, or difficulty walking. Because articular cartilage has a low self-repair ability and an extremely low proliferative capacity, healing of damaged articular cartilage has not been achieved to date. The current pharmaceutical treatment of OA is limited to the slight alleviation of symptoms (e.g., local injection of hyaluronic acid or non-steroidal anti-inflammatory drugs); hence, the development of effective drugs and regenerative therapies for OA is highly desirable. This review article summarizes findings indicating that proteoglycan 4 (Prg4)/lubricin, which is specifically expressed in the superficial zone of articular cartilage and synovium, functions in a protective manner against OA, and covers the transcriptional regulation of Prg4 in articular chondrocytes. We also focused on growth differentiation factor 5 (Gdf5), which is specifically expressed on the surface layer of articular cartilage, particularly in the developmental stage, describing its regulatory mechanisms and functions in joint formation and OA pathogenesis. Because several genetic studies in humans and mice indicate the involvement of these genes in the maintenance of articular cartilage homeostasis and the presentation of OA, molecular targeting of Prg4 and Gdf5 is expected to provide new insights into the aetiology, pathogenesis, and potential treatment of OA.
Collapse
|
52
|
Peripheral Blood-Derived Mesenchymal Stem Cells Modulate Macrophage Plasticity through the IL-10/STAT3 Pathway. Stem Cells Int 2022; 2022:5181241. [PMID: 35450344 PMCID: PMC9017453 DOI: 10.1155/2022/5181241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/23/2021] [Accepted: 03/14/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can skew the balance of M1/M2 macrophage polarization towards the M2 phenotype via their paracrine effects, thereby promoting anatomical and functional recovery after many inflammatory diseases induced by macrophages. However, the underlying mechanism is still poorly understood. This study focused on the IL-10/STAT3 pathway and investigated whether IL-10 secreted by PBMSCs could mediate M2 polarization through the activation of this pathway. In this study, a Transwell system was used for coculturing macrophages and PBMSCs. ELISA and RT-qPCR analysis found that PBMSCs and their conditioned media (P-CM) significantly induced the expression of IL-10, while significantly inhibiting the expression of IL-1β and TNF-α; moreover, this effect could be reversed by adding Ab9969 (an IL-10 neutralizing antibody) and Stattic (a STAT3 inhibitor). Furthermore, western blotting and immunofluorescence assays demonstrated that JAK1/STAT3 signaling was significantly upregulated in macrophages cocultured with PBMSCs or P-CM, accompanied by an increase in the M2 biomarker CD206 and a decrease in the M1 biomarker CD86. This effect could also be reversed by blocking the IL-10/STAT3 pathway with Ab9969 and Stattic. In summary, PBMSCs could mediate the polarization of M2 macrophages by activating the IL-10/STAT3 pathway.
Collapse
|
53
|
Zheng W, Li X, Li J, Wang X, Liu D, Zhai L, Ding B, Li G, Sun Y, Yokota H, Zhang P. Mechanical loading mitigates osteoarthritis symptoms by regulating the inflammatory microenvironment in a mouse model. Ann N Y Acad Sci 2022; 1512:141-153. [DOI: 10.1111/nyas.14760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/27/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Weiwei Zheng
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases Tianjin Medical University Tianjin China
| | - Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases Tianjin Medical University Tianjin China
| | - Xiaoyu Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases Tianjin Medical University Tianjin China
| | - Lidong Zhai
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Beibei Ding
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Guang Li
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Yuting Sun
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
| | - Hiroki Yokota
- Department of Biomedical Engineering Indiana University‐Purdue University Indianapolis Indianapolis Indiana
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences Tianjin Medical University Tianjin China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases Tianjin Medical University Tianjin China
- Tianjin Key Laboratory of Spine and Spinal Cord Tianjin Medical University Tianjin China
| |
Collapse
|
54
|
Wu Q, Sun S, Wei L, Liu M, Liu H, Liu T, Zhou Y, Jia Q, Wang D, Yang Z, Duan M, Yang X, Gao P, Ning X. Twist1 regulates macrophage plasticity to promote renal fibrosis through galectin-3. Cell Mol Life Sci 2022; 79:137. [PMID: 35182235 PMCID: PMC8858306 DOI: 10.1007/s00018-022-04137-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/21/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Renal interstitial fibrosis is the pathological basis of end-stage renal disease, in which the heterogeneity of macrophages in renal microenvironment plays an important role. However, the molecular mechanisms of macrophage plasticity during renal fibrosis progression remain unclear. In this study, we found for the first time that increased expression of Twist1 in macrophages was significantly associated with the severity of renal fibrosis in IgA nephropathy patients and mice with unilateral ureteral obstruction (UUO). Ablation of Twist1 in macrophages markedly alleviated renal tubular injury and renal fibrosis in UUO mice, accompanied by a lower extent of macrophage infiltration and M2 polarization in the kidney. The knockdown of Twist1 inhibited the chemotaxis and migration of macrophages, at least partially, through the CCL2/CCR2 axis. Twist1 downregulation inhibited M2 macrophage polarization and reduced the secretion of the profibrotic factors Arg-1, MR (CD206), IL-10, and TGF-β. Galectin-3 was decreased in the macrophages of the conditional Twist1-deficient mice, and Twist1 was shown to directly activate galectin-3 transcription. Up-regulation of galectin-3 recovered Twist1-mediated M2 macrophage polarization. In conclusion, Twist1/galectin-3 signaling regulates macrophage plasticity (M2 phenotype) and promotes renal fibrosis. This study could suggest new strategies for delaying kidney fibrosis in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Qingfeng Wu
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, No. 127 Chang le West Road, Xi'an, 710032, Shaanxi, China.,Department of Geriatrics, Ninth Hospital of Xi'an City, Xi'an, 710054, Shaanxi, China
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Lei Wei
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Minna Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Hao Liu
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Ting Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Ying Zhou
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Qing Jia
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Di Wang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhen Yang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Menglu Duan
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xiaoxia Yang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 2B. 71B, Baltimore, MD, 21224, USA.,Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
| | - Xiaoxuan Ning
- Department of Geriatrics, Xijing Hospital, Fourth Military Medical University, No. 127 Chang le West Road, Xi'an, 710032, Shaanxi, China. .,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
55
|
Nowaczyk A, Szwedowski D, Dallo I, Nowaczyk J. Overview of First-Line and Second-Line Pharmacotherapies for Osteoarthritis with Special Focus on Intra-Articular Treatment. Int J Mol Sci 2022; 23:1566. [PMID: 35163488 PMCID: PMC8835883 DOI: 10.3390/ijms23031566] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) can be defined as the result of pathological processes of various etiologies leading to damage to the articular structures. Although the mechanism of degenerative changes has become better understood due to the plethora of biochemical and genetic studies, the drug that could stop the degenerative cascade is still unknown. All available forms of OA therapy are based on symptomatic treatment. According to actual guidelines, comprehensive treatment of OA should always include a combination of various therapeutic options aimed at common goals, which are pain relief in the first place, and then the improvement of function. Local treatment has become more common practice, which takes place between rehabilitation and pharmacological treatment in the hierarchy of procedures. Only in the case of no improvement and the presence of advanced lesions visible in imaging tests, should surgery be considered. Currently, an increasing number of studies are being published suggesting that intra-articular injections may be as effective or even more effective than non-steroidal anti-inflammatory drugs (NSAIDs) and result in fewer systemic adverse events. The most commonly used preparations are hyaluronic acid (HA), glucocorticosteroids (GS), and also platelet-rich plasma (PRP) in recent years. This review aims to present the mechanism of action and clinical effectiveness of different pharmacological options in relieving pain and improving functions in OA as well as the emerging approach in intra-articular treatment with PRP.
Collapse
Affiliation(s)
- Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, LudwikRydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza St., 85-094 Bydgoszcz, Poland
| | - Dawid Szwedowski
- Department of Orthopaedics and Trauma Surgery, Provincial Polyclinical Hospital, 87-100 Toruń, Poland;
- Orthopedic Arthroscopic Surgery International (O.A.S.I.) Bioresearch Foundation, Gobbi N.P.O., 20133 Milan, Italy
| | - Ignacio Dallo
- Unit of Biological Therapies, SportMe Medical Center, Department of Orthopaedic Surgery and Sports Medicine, 41013 Seville, Spain;
| | - Jacek Nowaczyk
- Department of Physical Chemistry and Physicochemistry of Polymers, Faculty of Chemistry, Nicolaus Copernicus University, 7 Gagarina St., 87-100 Toruń, Poland
| |
Collapse
|
56
|
Li M, Li H, Ran X, Yin H, Luo X, Chen Z. Effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the osteoarthritis rabbit model. Arthritis Res Ther 2021; 23:294. [PMID: 34863246 PMCID: PMC8643028 DOI: 10.1186/s13075-021-02684-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/20/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The use of interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitor as a treatment for the inflammatory joint disease is a promising method. However, its underlying mechanism in osteoarthritis (OA) remains unclear. The purpose of this study is to look into the effects of adenovirus-mediated knockdown of IRAK4 on synovitis in the OA rabbit model. METHODS Ad-shIRAK4 was injected two weeks after anterior cruciate ligament resection. Six weeks later, the rabbits were killed. The expression of IRAK4, TNFR-associated factor 6(TRAF6), TGF-activated kinase 1(TAK1), p-IKB kinase (p-IKK), p-nuclear factor kappa-B (p-NFκB), p38, and p-p38 in the synovial membrane was detected by western blot, qRT-PCR, and immunohistochemistry analysis. Immunohistochemistry was to detect the expression of IRAK4 proteins in articular cartilage. H&E staining was to assess the pathological changes of synovium and cartilage. The levels of interleukin (IL)-1β, tumor necrosis factor-α(TNF-α), and MMP-13 in the synovial fluid were measured by ELISA. X-ray and micro-computerized tomography (μCT) scans were used to assess knee joint conditions and microstructure of subchondral bone. RESULTS IRAK4 expression levels in synovial tissues of the OA model group exhibited a significant upward trend. Ad-shIRAK4 significantly reduced IRAK4 mRNA expression in synovium tissues. Notably, Ad-shIRAK4 suppressed the Toll-like receptor/interleukin-1 receptor (TLR/IL-1R) signaling. In addition, in the Ad-shIRAK4 treatment group, we can see less inflammatory cell infiltration and reduced hyperplasia and angiogenesis. The levels of IL-1β, TNF-α, and MMP-13 in the synovial fluid in the OA model group were significantly higher than that in the control group, which were reduced by Ad-shIRAK4 treatment. Finally, Results of HE stains, immunohistochemistry, and μCT showed that Ad-shIRAK4 treatment has a protective effect on cartilage damage. CONCLUSIONS IRAK4 is significantly upregulated in the synovium from the osteoarthritis rabbit model. In addition, Ad-shIRAK4 reduced the expression of IRAK4 and suppressed TLR/IL-1R signaling in the synovium from the osteoarthritis rabbit model. Ad-shIRAK4 could alleviate synovitis and cartilage degradation in the osteoarthritis rabbit model, and thus alleviate the symptoms of OA and prevent the progression of OA.
Collapse
Affiliation(s)
- Muzhe Li
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Huiyun Li
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Xun Ran
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China
| | - Han Yin
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xuling Luo
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China.
| | - Zhiwei Chen
- Department of Orthopedic, The First Affiliated Hospital, Hengyang Medical School, University of South China, No. 69, Chuanshan Road, Hengyang City, 421001, Hunan Province, China.
| |
Collapse
|
57
|
Li J, Zhang Z, Qiu J, Huang X. 8-Methoxypsoralen has Anti-inflammatory and Antioxidant Roles in Osteoarthritis Through SIRT1/NF-κB Pathway. Front Pharmacol 2021; 12:692424. [PMID: 34552480 PMCID: PMC8450503 DOI: 10.3389/fphar.2021.692424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022] Open
Abstract
Osteoarthritis (OA) is mainly manifested by joint pain, stiffness and mobility disorder, which is the main cause of pain and disability in middle-aged and elderly people. In this study, we aimed to explore the role and mechanism of 8-Methoxypsoralen (8-MOP) in the OA model both in vitro and in vivo. The rat chondrocytes were treated with IL-1β, and the proliferation, apoptosis, inflammatory reactions and oxidative stress responses were determined after treatment with different concentrations of 8-MOP. Real-time quantitative polymerase chain reaction (qRT-PCR) and/or Western blot were implemented to check the AMPK/SIRT1/NF-κB expression in chondrocytes. The NF-κB activity was determined by dual luciferase experiment. The pain threshold of OA rat model dealt with 8-MOP and/or the SIRT1 inhibitor EX527 was measured. Our results revealed that 8-MOP evidently reduced IL-1β-mediated apoptosis and inhibition of proliferation, and mitigated the expression of inflammatory cytokines and oxidative stress factors in chondrocytes. Additionally, 8-MOP promoted phosphorylated level of AMPKα, enhanced SIRT1 expression and inhibited the phosphorylation of NF-κB. After treatment with EX527, 8-MOP-mediated protective effects on chondrocytes were mostly reversed. In vivo, 8-MOP obviously improved the pain threshold in the OA rat model and reduced the injury and apoptosis of chondrocytes in the joints. In addition, 8-MOP relieved inflammatory and oxidative stress responses in the articular cartilage via enhancing SIRT1 and repressing NF-κB activation. After the treatment with EX527, the 8-MOP-mediated protective effects were distinctly weakened. In summary, our study testified that 8-MOP alleviates pain, inflammatory and oxidative stress responses in OA rats through the SIRT1/NF-κB pathway, which is expected to become a new reagent for clinical treatment of OA.
Collapse
Affiliation(s)
- Jichao Li
- The Third Department of Knee Injury, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Zeng Zhang
- Zhengzhou Orthopedic Hospital, Zhengzhou, China
| | - Jinan Qiu
- The Third Department of Knee Injury, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| | - Xiaohan Huang
- The Third Department of Knee Injury, Luoyang Orthopedic Hospital of Henan Province, Orthopedic Hospital of Henan Province, Luoyang, China
| |
Collapse
|
58
|
Attenuation of Knee Osteoarthritis Progression in Mice through Polarization of M2 Macrophages by Intra-Articular Transplantation of Non-Cultured Human Adipose-Derived Regenerative Cells. J Clin Med 2021; 10:jcm10194309. [PMID: 34640324 PMCID: PMC8509129 DOI: 10.3390/jcm10194309] [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: 08/15/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
Adipose-derived regenerative cells (ADRCs) are non-cultured heterogeneous or mixed populations of cells obtained from adipose tissue by collagenase digestion. The injection of ADRCs have been tried clinically for the treatment of osteoarthritis (OA). The purpose of this study was to evaluate the effect of intra-articular transplantation of human ADRCs on OA progression in mice and the effect of ADRCs on macrophage polarization. In in vivo experiments, BALB/c-nu mice with knee OA received intra-articular transplantation of either phosphate buffered-saline or human ADRCs. OA progression was evaluated histologically and significantly attenuated in the ADRC group at both four and eight weeks postoperatively. The expression of OA-related proteins in the cartilage and macrophage-associated markers in the synovium were examined by immunohistochemistry. The numbers of MMP-13-, ADAMTS-5-, IL-1β-, IL-6- and iNOS-positive cells significantly decreased, and type II collagen- and CD206-positive cells were more frequently detected in the ADRC group compared with that in the control group. In vitro co-culture experiments showed that ADRCs induced macrophage polarization toward M2. The results of this study suggest that the intra-articular transplantation of human ADRCs could attenuate OA progression possibly by reducing catabolic factors in chondrocytes and modulating macrophage polarization.
Collapse
|
59
|
Shojarazavi N, Mashayekhan S, Pazooki H, Mohsenifard S, Baniasadi H. Alginate/cartilage extracellular matrix-based injectable interpenetrating polymer network hydrogel for cartilage tissue engineering. J Biomater Appl 2021; 36:803-817. [PMID: 34121491 DOI: 10.1177/08853282211024020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the present study, alginate/cartilage extracellular matrix (ECM)-based injectable hydrogel was developed incorporated with silk fibroin nanofibers (SFN) for cartilage tissue engineering. The in situ forming hydrogels were composed of different ionic crosslinked alginate concentrations with 1% w/v enzymatically crosslinked phenolized cartilage ECM, resulting in an interpenetrating polymer network (IPN). The response surface methodology (RSM) approach was applied to optimize IPN hydrogel's mechanical properties by varying alginate and SFN concentrations. The results demonstrated that upon increasing the alginate concentration, the compression modulus improved. The SFN concentration was optimized to reach a desired mechanical stiffness. Accordingly, the concentrations of alginate and SFN to have an optimum compression modulus in the hydrogel were found to be 1.685 and 1.724% w/v, respectively. The gelation time was found to be about 10 s for all the samples. Scanning electron microscope (SEM) images showed homogeneous dispersion of the SFN in the hydrogel, mimicking the natural cartilage environment. Furthermore, water uptake capacity, degradation rate, cell cytotoxicity, and glycosaminoglycan and collagen II secretions were determined for the optimum hydrogel to support its potential as an injectable scaffold for articular cartilage defects.
Collapse
Affiliation(s)
- Nastaran Shojarazavi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Shohreh Mashayekhan
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hossein Pazooki
- Department of Chemical and Environmental Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Sadaf Mohsenifard
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hossein Baniasadi
- Polymer Technology, School of Chemical Engineering, Aalto University, Espoo, Finland
| |
Collapse
|
60
|
Zhang M, Zhang R, Zheng T, Chen Z, Ji G, Peng F, Wang W. Xanthohumol Attenuated Inflammation and ECM Degradation by Mediating HO-1/C/EBPβ Pathway in Osteoarthritis Chondrocytes. Front Pharmacol 2021; 12:680585. [PMID: 34017261 PMCID: PMC8129538 DOI: 10.3389/fphar.2021.680585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 04/23/2021] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is the most frequent and disabling disease in developed countries. The progressive degeneration of articular cartilage characterized as thinner and erosive. Inflammation is well-known to be involved in OA development. However, there are no effective therapeutic strategies to cure it. Xanthohumol (XH) is a natural prenylflavonoid isolated from hops and beer. The protective activity of XH against OA chondrocytes inflammation and ECM degradation is unclear. In this article, we found that XH significantly inhibited inflammatory responses, attenuated catabolic enzymes expression, and ameliorated ECM degradation, as showed by decreased production of NO, PGE2, TNFα, and IL-6, decreased expression of MMP-3/-13 and ADAMTS-4/-5, and increased expression of collagen-II and aggrecan. In addition, XH activated HO-1 signaling and attenuated IL-1β-induced C/EBPβ. XH promoted the interaction between HO-1 and C/EBPβ, inhibiting the nuclear translocation of C/EBPβ. HO-1 knockdown could abrogate the protective effects of XH in IL-1β-treated chondrocytes. Collectively, XH attenuated inflammatory responses and ECM degradation by mediating HO-1 and C/EBPβ signaling pathways in osteoarthritis chondrocytes.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Orthopedics, Taizhou People's Hospital, Taizhou, China
| | - Rui Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Tiansheng Zheng
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Guanglin Ji
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fang Peng
- Department of Pathology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou, China
| | - Wei Wang
- Department of Hepatology, Taizhou People's Hospital, Taizhou, China
| |
Collapse
|
61
|
Li PL, Wang YX, Zhao ZD, Li ZL, Liang JW, Wang Q, Yin BF, Hao RC, Han MY, Ding L, Wu CT, Zhu H. Clinical-grade human dental pulp stem cells suppressed the activation of osteoarthritic macrophages and attenuated cartilaginous damage in a rabbit osteoarthritis model. Stem Cell Res Ther 2021; 12:260. [PMID: 33933140 PMCID: PMC8088312 DOI: 10.1186/s13287-021-02353-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/23/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Although increasing evidence has demonstrated that human dental pulp stem cells (hDPSCs) are efficacious for the clinical treatment of skeletal disorders, the underlying mechanisms remain incompletely understood. Osteoarthritis (OA) is one of the most common degenerative disorders in joints and is characterized by gradual and irreversible cartilaginous tissue damage. Notably, immune factors were newly identified to be closely related to OA development. In this study, we explored the modulatory effects of clinical-grade hDPSCs on osteoarthritic macrophages and their protective effects on cartilaginous tissues in OA joints. METHODS The cell morphology, immunophenotype, and inflammatory factor expression of osteoarthritic macrophages were explored by phase contrast microscope, transmission electron microscopy, immunostaining, flow cytometry, quantitative polymerase chain reaction, and enzyme linked immunosorbent assay, respectively. Additionally, the factors and signaling pathways that suppressed macrophage activation by hDPSCs were determined by enzyme-linked immunosorbent assay and western-blotting. Furthermore, hDPSCs were administered to a rabbit knee OA model via intra-articular injection. Macrophage activation in vivo and cartilaginous tissue damage were also evaluated by pathological analysis. RESULTS We found that hDPSCs markedly inhibited osteoarthritic macrophage activation in vitro. The cell morphology, immunophenotype, and inflammatory factor expression of osteoarthritic macrophages changed into less inflammatory status in the presence of hDPSCs. Mechanistically, we observed that hDPSC-derived hepatocyte growth factor and transforming growth factor β1 mediated the suppressive effects on osteoarthritic macrophages. Moreover, phosphorylation of MAPK pathway proteins contributed to osteoarthritic macrophage activation, and hDPSCs suppressed their activation by partially inactivating those pathways. Most importantly, injected hDPSCs inhibited macrophage activation in osteochondral tissues in a rabbit knee OA model in vivo. Further histological analysis showed that hDPSCs alleviated cartilaginous damage to knee joints. CONCLUSIONS In summary, our findings reveal a novel function for hDPSCs in suppressing osteoarthritic macrophages and suggest that macrophages are efficient cellular targets of hDPSCs for alleviation of cartilaginous damage in OA. hDPSCs treat OA via an osteoarthritic macrophages-dependent mechanisms. hDPSCs suppress the activation of osteoarthritic macrophages in vitro and in vivo and alleviate cartilaginous lesions in OA models.
Collapse
Affiliation(s)
- Pei-Lin Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Yu-Xing Wang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, People's Republic of China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Zhi-Dong Zhao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Zhi-Ling Li
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Jia-Wu Liang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Qian Wang
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- People's Liberation Army General Hospital, Road Fuxing 28, Beijing, 100853, P. R. China
| | - Bo-Feng Yin
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Rui-Cong Hao
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
- Graduate School of Anhui Medical University, 81 Meishan Road, Shu Shan Qu, Hefei, 230032, Anhui, P. R. China
| | - Meng-Yue Han
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
- Graduate School of Anhui Medical University, 81 Meishan Road, Shu Shan Qu, Hefei, 230032, Anhui, P. R. China
| | - Li Ding
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China.
- Medical Center of Air Forces, PLA, Road Fucheng 30, Beijing, 100142, P. R. China.
| | - Chu-Tse Wu
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China.
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, Road Taiping 27, Beijing, 100850, P. R. China.
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
- Graduate School of Anhui Medical University, 81 Meishan Road, Shu Shan Qu, Hefei, 230032, Anhui, P. R. China.
| |
Collapse
|
62
|
Wang H, Jiang Z, Pang Z, Qi G, Hua B, Yan Z, Yuan H. Engeletin Protects Against TNF-α-Induced Apoptosis and Reactive Oxygen Species Generation in Chondrocytes and Alleviates Osteoarthritis in vivo. J Inflamm Res 2021; 14:745-760. [PMID: 33727849 PMCID: PMC7955871 DOI: 10.2147/jir.s297166] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
Purpose Osteoarthritis (OA) is a progressive disease characterized by pain and impaired joint functions. Engeletin is a natural compound with anti-inflammatory and antioxidant effects on other diseases, but the effect of engeletin on OA has not been evaluated. This study aimed to elucidate the protective effect of engeletin on cartilage and the underlying mechanisms. Methods Chondrocytes were isolated from rat knee cartilage, and TNF-α was used to simulate OA in vitro. After treatment with engeletin, the expression of extracellular matrix (ECM) components (collagen II and aggrecan) and matrix catabolic enzymes (MMP9 and MMP3) was determined by Western blotting and qPCR. Chondrocyte apoptosis was evaluated using Annexin V-FITC/PI and flow cytometry. Apoptosis-related proteins (Bax, Bcl-2, and cleaved caspase-3) were evaluated by Western blotting. The mitochondrial membrane potential of chondrocytes was measured with JC-1, and intracellular reactive oxygen species (ROS) levels were determined with DCFH-DA. Changes in signaling pathways (Nrf2, NF-κB and MAPK) were evaluated by Western blotting. In vivo, anterior cruciate ligament transection (ACLT) was used to induce the rat OA model, and engeletin was administered intraarticularly. The therapeutic effect of engeletin was analyzed by histopathological analysis. Results Pretreatment with engeletin alleviated TNF-α-induced inhibition of ECM components (collagen II and aggrecan) and upregulation of matrix catabolic enzymes (MMP9 and MMP3). Engeletin ameliorated chondrocyte apoptosis by inhibiting Bax expression and upregulating Bcl-2 expression. Engeletin maintained the mitochondrial membrane potential of chondrocytes and scavenged intracellular ROS by activating the Nrf2 pathway. The NF-κB and MAPK pathways were inhibited by treatment with engeletin. In vivo, ACLT-induced knee OA in rats was alleviated by intraarticular injection of engeletin. Conclusion Engeletin ameliorated OA in vitro and in vivo. It may be a potential therapeutic drug for OA.
Collapse
Affiliation(s)
- Hao Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zengxin Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zhiying Pang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Guobin Qi
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Bingxuan Hua
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Zuoqin Yan
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Hengfeng Yuan
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, People's Republic of China
| |
Collapse
|