51
|
Pastrello C, Kotlyar M, Abovsky M, Lu R, Jurisica I. PathDIP 5: improving coverage and making enrichment analysis more biologically meaningful. Nucleic Acids Res 2024; 52:D663-D671. [PMID: 37994706 PMCID: PMC10767947 DOI: 10.1093/nar/gkad1027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/24/2023] Open
Abstract
Pathway Data Integration Portal (PathDIP) is an integrated pathway database that was developed to increase functional gene annotation coverage and reduce bias in pathway enrichment analysis. PathDIP 5 provides multiple improvements to enable more interpretable analysis: users can perform enrichment analysis using all sources, separate sources or by combining specific pathway subsets; they can select the types of sources to use or the types of pathways for the analysis, reducing the number of resulting generic pathways or pathways not related to users' research question; users can use API. All pathways have been mapped to seven representative types. The results of pathway enrichment can be summarized through knowledge-based pathway consolidation. All curated pathways were mapped to 53 pathway ontology-based categories. In addition to genes, pathDIP 5 now includes metabolites. We updated existing databases, included two new sources, PathBank and MetabolicAtlas, and removed outdated databases. We enable users to analyse their results using Drugst.One, where a drug-gene network is created using only the user's genes in a specific pathway. Interpreting the results of any analysis is now improved by multiple charts on all the results pages. PathDIP 5 is freely available at https://ophid.utoronto.ca/pathDIP.
Collapse
Affiliation(s)
- Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, Toronto, Ontario M5T 0S8, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON M5T 0S8, Canada
| | - Max Kotlyar
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, Toronto, Ontario M5T 0S8, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON M5T 0S8, Canada
| | - Mark Abovsky
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, Toronto, Ontario M5T 0S8, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON M5T 0S8, Canada
| | - Richard Lu
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, Toronto, Ontario M5T 0S8, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON M5T 0S8, Canada
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, Toronto, Ontario M5T 0S8, Canada
- Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON M5T 0S8, Canada
- Departments of Medical Biophysics and Computer Science, and Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1L7, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| |
Collapse
|
52
|
Lv X, Wang X, Wang X, Han Y, Chen H, Hao Y, Zhang H, Cui C, Gao Q, Zheng Z. Research progress in arthritis treatment with the active components of Herba siegesbeckiae. Biomed Pharmacother 2023; 169:115939. [PMID: 38007937 DOI: 10.1016/j.biopha.2023.115939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023] Open
Abstract
Arthritis is a group of diseases characterized by joint pain, swelling, stiffness, and limited movement. Osteoarthritis, rheumatoid arthritis, and gouty arthritis are the most common types of arthritis. Arthritis severely affects the quality of life of patients and imposes a heavy financial and medical burden on their families and society at large. As a widely used traditional Chinese medicine, Herba siegesbeckiae has many pharmacological effects such as anti-inflammatory and analgesic, anti-ischemic injury, cardiovascular protection, and hypoglycemic. In addition, it has significant therapeutic effects on arthritis. The rich chemical compositions of H. siegesbeckiae primarily include diterpenoids, sesquiterpenoids, and flavonoids. As one of the main active components of H. siegesbeckiae, kirenol and quercetin play a vital role in reducing arthritis symptoms. In the present study, the research progress in arthritis treatment with the active components of H. siegesbeckiae is reviewed.
Collapse
Affiliation(s)
- Xiaoqian Lv
- Binzhou Medical University, 264003 Yantai, China
| | - Xiaoyu Wang
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China
| | - Xuelei Wang
- Binzhou Medical University, 264003 Yantai, China
| | - Yunna Han
- Binzhou Medical University, 264003 Yantai, China
| | - Haoyue Chen
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China
| | - Yuwen Hao
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China
| | - Hao Zhang
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China
| | - Chao Cui
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China
| | - Qiang Gao
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China.
| | - Zuncheng Zheng
- The Affiliated Taian City Central Hospital of Qingdao University, 271000 Taian, China.
| |
Collapse
|
53
|
Qiao S, Zhao R, He S, Fu X, An J, Xia T. Quercitrin attenuates the progression of osteoarthritis via inhibiting NF-κB signaling pathways and enhance glucose transport capacity. Exp Cell Res 2023; 433:113854. [PMID: 37952573 DOI: 10.1016/j.yexcr.2023.113854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Osteoarthritis (OA) is a common musculoskeletal disorder that impairs function and reduces the quality of life. Extracellular matrix (ECM) degradation and inflammatory mechanisms are crucial to the progression of OA. In this study, we aimed to investigate the anti-inflammatory activity, anti-ECM degradation property, and glucose transport capacity of quercitrin (QCT) on IL-1β-treated rat primary chondrocytes. Rat primary chondrocytes were treated with IL-1β to simulate inflammatory environmental conditions and OA in vitro. We examined the effects of QCT at concentrations ranging from 0 to 200 μM on the viability of rat chondrocytes and selected 5 μM for further study. Using qRT-PCR, immunofluorescent, immunocytochemistry, and western blotting techniques, we identified the potential molecular mechanisms and signaling pathways that are responsible for these effects. We established an OA rat model through anterior cruciate ligament transection (ACLT). The animals were then periodically injected with QCT into the knee articular cavity. Our in vivo and in vitro study showed that QCT could inhibit IL-1β-activated inflammation and ECM degradation in chondrocyte. Furthermore, QCT could inhibit the NF-κB signal pathway and enhance glucose transport capacity in the IL-1β-stimulated chondrocytes. In vivo study proved that QCT attenuates OA progression in rats. Overall, QCT inhibited the activation of NF-κB and enhanced glucose transport capacity to alleviate the progression of OA.
Collapse
Affiliation(s)
- Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Runze Zhao
- Department of Orthopedic Surgery, The First Affiliated Hospital, Orthopedic Institure, Medical College, Soochow University, Suzhou, China
| | - Shuangjian He
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Xuejie Fu
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Tingting Xia
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China.
| |
Collapse
|
54
|
Sun J, Chen W, Zhou Z, Chen X, Zuo Y, He J, Liu H. Tanshinone IIA Facilitates Efficient Cartilage Regeneration under Inflammatory Factors Caused Stress via Upregulating LncRNA NEAT1_2. Biomedicines 2023; 11:3291. [PMID: 38137512 PMCID: PMC10741062 DOI: 10.3390/biomedicines11123291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/29/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
(1) Background: Osteoarthritis (OA) is a crippling condition characterized by chondrocyte dedifferentiation, cartilage degradation, and subsequent cartilage defects. Unfortunately, there is a lack of effective medicines to facilitate the repair of cartilage defects in OA patients. In this study, we investigated the role of lncRNA NEAT1_2 in maintaining the chondrocyte phenotype and identified tanshinone IIA(TAN) as a natural medicine that enhances NEAT1_2 levels, resulting in efficient cartilage regeneration under inflammatory cytokines. (2) Methods: The transcriptional levels of NEAT1_2 and cartilage phenotype-related genes were identified by RT-qPCR. The siRNA interference approach was utilized to silence NEAT1_2; the Alamar Blue assay was performed to determine chondrocyte viability under inflammatory conditions. To evaluate the concentrations of collagen type II and glycosaminoglycans distributed by chondrocytes in vitro and in vivo, immunohistochemical staining and Safranin O staining were used. (3) Results: IL-1β suppresses NEAT1_2 and genes related to the chondrocytic phenotype, whereas TAN effectively upregulates them in a NEAT1_2-dependent manner. Consistently, TAN alleviated chondrocyte oxidative stress inhibited cartilage degradation by modulating the relevant genes and promoted efficient cartilage regeneration in vitro and in vivo when chondrocytes are exposed to inflammatory cytokines. (4) Conclusions: TAN enhances the expression of NEAT1_2 inhibited by IL-1β and affects the transcription of chondrocytic phenotype-related genes, which promotes cartilage regeneration in an inflammatory environment.
Collapse
Affiliation(s)
- Jingjing Sun
- College of Biology, Hunan University, Changsha 410082, China; (J.S.); (Y.Z.); (J.H.)
| | - Wei Chen
- College of Material Science and Engineering, Hunan University, Changsha 410082, China; (W.C.); (X.C.)
| | - Zheng Zhou
- College of Biology, Hunan University, Changsha 410082, China; (J.S.); (Y.Z.); (J.H.)
| | - Xin Chen
- College of Material Science and Engineering, Hunan University, Changsha 410082, China; (W.C.); (X.C.)
| | - You Zuo
- College of Biology, Hunan University, Changsha 410082, China; (J.S.); (Y.Z.); (J.H.)
| | - Jiaqian He
- College of Biology, Hunan University, Changsha 410082, China; (J.S.); (Y.Z.); (J.H.)
| | - Hairong Liu
- College of Material Science and Engineering, Hunan University, Changsha 410082, China; (W.C.); (X.C.)
| |
Collapse
|
55
|
Chen H, Li Z, Li X, Lu J, Chen B, Wang Q, Wu G. Biomaterial-Based Gene Delivery: Advanced Tools for Enhanced Cartilage Regeneration. Drug Des Devel Ther 2023; 17:3605-3624. [PMID: 38076630 PMCID: PMC10706074 DOI: 10.2147/dddt.s432056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
Gene therapy has emerged as a promising and innovative approach in cartilage regeneration. Integrating biomaterials into gene therapy offers a unique opportunity to enhance gene delivery efficiency, optimize gene expression dynamics, modulate immune responses, and promote tissue regeneration. Despite the rapid progress in biomaterial-based gene delivery, there remains a deficiency of comprehensive discussions on recent advances and their specific application in cartilage regeneration. Therefore, this review aims to provide a thorough overview of various categories of biomaterials employed in gene delivery, including both viral and non-viral vectors, with discussing their distinct advantages and limitations. Furthermore, the diverse strategies employed in gene therapy are discussed and summarized, such as the utilization of growth factors, anti-inflammatory cytokines, and chondrogenic genes. Additionally, we highlights the significant challenges that hinder biomaterial-based gene delivery in cartilage regeneration, including immune response modulation, gene delivery efficiency, and the sustainability of long-term gene expression. By elucidating the functional properties of biomaterials-based gene therapy and their pivotal roles in cartilage regeneration, this review aims to enhance further advances in the design of sophisticated gene delivery systems for improved cartilage regeneration outcomes.
Collapse
Affiliation(s)
- Hongfeng Chen
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Zhen Li
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Xiaoqi Li
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Jiongjiong Lu
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Beibei Chen
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Qiongchao Wang
- Department of Foot and Ankle Surgery, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| | - Guangliang Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462300, People’s Republic of China
| |
Collapse
|
56
|
Cao N, Wang D, Liu B, Wang Y, Han W, Tian J, Xiang L, Wang Z. Silencing of STUB1 relieves osteoarthritis via inducing NRF2-mediated M2 macrophage polarization. Mol Immunol 2023; 164:112-122. [PMID: 37992540 DOI: 10.1016/j.molimm.2023.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVES Shifting macrophages towards an anti-inflammatory state is key in treating osteoarthritis (OA) by reducing inflammation and tissue damage. However, the underlying mechanisms guiding this shift remain largely undefined. STUB1, an E3 ubiquitin ligase, known for its regulatory role in macrophage polarization. This study aims to explore the function and underlying action mechanisms of STUB1 in OA. METHODS An in vivo OA model was established in rats. Hematoxylin-Eosin and safranin O-fast green staining were performed to reveal the hispathological injuries in knee-joint tissues. Immunohistochemistry and flow cytometry were performed to detect the distribution of M1 and M2 macrophages. The inflammatory response (TNF-α and IL-6 levels) was evaluated by ELISA. In vitro, the interaction between STUB1 and NFR2 was determined by CO-IP and pull-down assays. After treated with LPS (an in vitro model of OA), the viability and apoptosis of chondrocytes were measured by CCK-8 and flow cytometry, respectively. RESULTS Silencing STUB1 alleviated OA in rats, as indicated by reduced subchondral bone thickness, knee synovitis score, histopathological damages, and inflammatory response. STUB1 silencing also decreased M1 macrophages and increased M2 macrophages in both in vivo and in vitro settings. NRF2 was identified as a target of STUB1, with STUB1 mediating its ubiquitination. Silencing NRF2 reversed the effects of STUB1 silencing on inducing M2 macrophage polarization. Furthermore, silencing STUB1 upregulated NRF2 expression in LPS-treated chondrocytes, promoting cell viability and inhibiting apoptosis. CONCLUSION Silencing STUB1 induces M2 macrophage polarization by inhibiting NRF2 ubiquitination, thereby contributing to the mitigation of OA.
Collapse
Affiliation(s)
- Nan Cao
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Danni Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Bin Liu
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Yu Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Wenfeng Han
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Jing Tian
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China.
| | - Zheng Wang
- Department of Orthopedics, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning Province, China.
| |
Collapse
|
57
|
Zhou M, Qiang J, Gan J, Xu X, Li X, Zhang S, Xu B, Dong Z. Quercetin attenuates environmental Avermectin-induced ROS accumulation and alleviates gill damage in carp through activation of the Nrf2 pathway. Comp Biochem Physiol C Toxicol Pharmacol 2023; 274:109744. [PMID: 37704162 DOI: 10.1016/j.cbpc.2023.109744] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/24/2023] [Accepted: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Avermectin (AVM) is one of the most often used insecticides which is toxic to aquatic organisms, and cause oxidative-induced damages to the fish respiratory organ, the "gills". To better understand the mechanism by which an antioxidant reduces AVM-induced gill damage, we investigated the effects of Quercetin (Que) on AVM induction of oxidative stress to inhibit damages to the gills using common carp as a model organism. The Que is a fruit and vegetable rich flavonoid with antioxidant activity. In this study, four groups were created: the Control group, the Que group (400 mg/kg), the AVM group (2.404 μg/L), and the Que plus AVM group. The analytical methods were pathological structure examination, qPCR, Reactive Oxygen Species (ROS) and Western blot. The results showed that Que alleviated AVM-induced oxidative stress, inflammatory damage and apoptosis in the carp gills by activating the Nrf2 pathway. The mechanism was that Que alleviated the accumulation of ROS, reduced the balance between oxidation and antioxidant disrupted by AVM exposure, lowered the content of lipid peroxidation produced malondialdehyde (MDA), and increased the content of antioxidant enzymes including glutathione (GSH) and catalase (CAT). Nrf2 pathway was activated. Meanwhile, Que inhibited gill apoptosis in carp by decreasing the levels of Bax, Cytochrome C, Caspase9, Cleaved-Caspase3 and reduced Bcl2. This has important implications for future studies on Que and AVM. New suggestions are provided to reduce the threat of aquatic environmental pollution.
Collapse
Affiliation(s)
- Mengyuan Zhou
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jingchao Qiang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jiajie Gan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xuhui Xu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xing Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Shuai Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Baoshi Xu
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zibo Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
58
|
Xu R, Kuang M, Li N. Phytochemistry and pharmacology of plants in the genus Chaenomeles. Arch Pharm Res 2023; 46:825-854. [PMID: 38062238 DOI: 10.1007/s12272-023-01475-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/25/2023] [Indexed: 12/17/2023]
Abstract
Chaenomeles plants belong to the Rosaceae family and include five species, Chaenomeles speciosa (Sweet) Nakai, Chaenomeles sinensis (Thouin) Koehne, Chaenomeles japonica (Thunb.) Lindl, Chaenomeles cathayensis (Hemsl.) Schneid and Chaenomeles thibetica Yu. Chaenomeles plants are found and cultivated in nearly every country worldwide. China serves as both the origin and distribution hub for the plants in the Chaenomeles genus, and all Chaenomeles species except for C. japonica are indigenous to China. Chaenomeles spp. is a type of edible medicinal plant that has been traditionally used in China to treat various ailments, such as rheumatism, cholera, dysentery, enteritis, beriberi, and scurvy. A variety of chemical constituents have been extracted from this genus, including terpenoids, phenolics, flavonoids, phenylpropanoids and their derivatives, benzoic acid derivatives, biphenyls, oxylipins, and alkaloids. The biological activity of some of these constituents has already been evaluated. Pharmacological investigations have demonstrated that the plants in the genus Chaenomeles exhibit anti-inflammatory, analgesic, antioxidant, antihyperglycemic, antihyperlipidemic, gastrointestinal protective, antitumor, immunomodulatory, antibacterial, antiviral, hepatoprotective, neuroprotective and other pharmacological activities. The objective of this review is to provide a comprehensive and up-to-date summary of the available information on the genus Chaenomeles to serve as a valuable reference for further investigations.
Collapse
Affiliation(s)
- Ruoling Xu
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Mengting Kuang
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Ning Li
- Anhui Key Laboratory of Bioactivity of Natural Products, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
59
|
Saha S, Rebouh NY. Anti-Osteoarthritis Mechanism of the Nrf2 Signaling Pathway. Biomedicines 2023; 11:3176. [PMID: 38137397 PMCID: PMC10741080 DOI: 10.3390/biomedicines11123176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease and the primary pathogenic consequence of OA is inflammation, which can affect a variety of tissues including the synovial membrane, articular cartilage, and subchondral bone. The development of the intra-articular microenvironment can be significantly influenced by the shift of synovial macrophages between pro-inflammatory and anti-inflammatory phenotypes. By regulating macrophage inflammatory responses, the NF-κB signaling route is essential in the therapy of OA; whereas, the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway appears to manage the relationship between oxidative stress and inflammation. Additionally, it has been demonstrated that under oxidative stress and inflammation, there is a significant interaction between transcriptional pathways involving Nrf2 and NF-κB. Studying how Nrf2 signaling affects inflammation and cellular metabolism may help us understand how to treat OA by reprogramming macrophage behavior because Nrf2 signaling is thought to affect cellular metabolism. The candidates for treating OA by promoting an anti-inflammatory mechanism by activating Nrf2 are also reviewed in this paper.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Nazih Y. Rebouh
- Department of Environmental Management, Institute of Environmental Engineering, RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russia
| |
Collapse
|
60
|
Zhang C, Zhou X, Wang D, Hao L, Zeng Z, Su L. Hydrogel-Loaded Exosomes: A Promising Therapeutic Strategy for Musculoskeletal Disorders. J Clin Pharm Ther 2023; 2023:1-36. [DOI: 10.1155/2023/1105664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Clinical treatment strategies for musculoskeletal disorders have been a hot research topic. Accumulating evidence suggests that hydrogels loaded with MSC-derived EVs show great potential in improving musculoskeletal injuries. The ideal hydrogels should be capable of promoting the development of new tissues and simulating the characteristics of target tissues, with the properties matching the cell-matrix constituents of autologous tissues. Although there have been numerous reports of hydrogels loaded with MSC-derived EVs for the repair of musculoskeletal injuries, such as intervertebral disc injury, tendinopathy, bone fractures, and cartilage injuries, there are still many hurdles to overcome before the clinical application of modified hydrogels. In this review, we focus on the advantages of the isolation technique of EVs in combination with different types of hydrogels. In this context, the efficacy of hydrogels loaded with MSC-derived EVs in different musculoskeletal injuries is discussed in detail to provide a reference for the future application of hydrogels loaded with MSC-derived EVs in the clinical treatment of musculoskeletal injuries.
Collapse
Affiliation(s)
- Chunyu Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Xuchang Zhou
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
| | - Li Hao
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Zhipeng Zeng
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing 100084, China
- Shougang Technician College, Nursing School, Beijing 100043, China
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| | - Lei Su
- Department of Rehabilitation, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou 510000, China
| |
Collapse
|
61
|
Bhoi A, Dwivedi SD, Singh D, Keshavkant S, Singh MR. Mechanistic prospective and pharmacological attributes of quercetin in attenuation of different types of arthritis. 3 Biotech 2023; 13:362. [PMID: 37840879 PMCID: PMC10570262 DOI: 10.1007/s13205-023-03787-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Arthritis is a frequent autoimmune disease with undefined etiology and pathogenesis. Scientific community constantly fascinating quercetin (QUR), as it is the best-known flavonoid among others for curative and preventive properties against a wide range of diseases. Due to its multifaceted activities, the implementation of QUR against various types of arthritis namely, rheumatoid arthritis (RA), osteoarthritis (OA), gouty arthritis (GA) and psoriotic arthritis (PsA) has greatly increased in recent years. Many research evidenced that QUR regulates a wide range of pathways for instance NF-κB, MAK, Wnt/β-catenine, Notch, etc., that are majorly associated with the inflammatory mechanisms. Besides, the bioavailability of QUR is a major constrain to its therapeutic potential, and drug delivery techniques have experienced significant development to overcome the problem of its limited application. Hence, this review compiled the cutting-edge experiments on versatile effects of QUR on inflammatory diseases like RA, OA, GA and PsA, sources and bioavailability, therapeutic challenges, pharmacokinetics, clinical studies as well as toxicological impacts. The use of QUR in a health context would offer a tearing and potential therapeutic method, supporting the advancement of public health, particularly, of arthritic patients worldwide.
Collapse
Affiliation(s)
- Anita Bhoi
- School of Studies in Biotechnology, Pt. Ravishankar Shukla University, Raipur, 492 010 India
| | - Shradha Devi Dwivedi
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010 India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010 India
| | - S. Keshavkant
- School of Studies in Biotechnology, Pt. Ravishankar Shukla University, Raipur, 492 010 India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010 India
| |
Collapse
|
62
|
Li X, Tao H, Zhou J, Zhang L, Shi Y, Zhang C, Sun W, Chu M, Chen K, Gu C, Yang X, Geng D, Hao Y. MAGL inhibition relieves synovial inflammation and pain via regulating NOX4-Nrf2 redox balance in osteoarthritis. Free Radic Biol Med 2023; 208:13-25. [PMID: 37516370 DOI: 10.1016/j.freeradbiomed.2023.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/31/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage injury, hyperplasia of bone and inflammatory lesions of synovium. Monoacylglycerol lipase (MAGL), a member of the α/β hydrolase superfamily, is involved in regulation of injury protection and immune-inflammation response. Autoinflammatory response of the synovium and the release of inflammatory mediators play critical roles in occurrence of early-stage OA. Fibroblast-like synoviocytes (FLSs) are resident mesenchymal cells of the synovial tissue. Considering that MAGL inhibition regulates the inflammatory signaling cascade, it is crucial to ascertain the biological effects and specific mechanisms of MAGL in alleviating inflammatory infiltration of OA FLSs. The aim of this study was to investigate the effect of MAGL on biological function in OA FLSs. Results from in vitro experiments showed that MAGL blockade not only effectively inhibited proliferation, invasion and migration of FLSs, but also downregulated expression of inflammatory-associated proteins. Sequencing results indicated that MAGL inhibition significantly suppressed NOX4-mediated oxidative stress, thus promoting Nrf2 nuclear accumulation and inhibiting generation of intracellular reactive oxygen species (ROS). Attenuation of NOX4 further alleviated redox dysplasia and ultimately improved tumor-like phenotypes, such as abnormal proliferation, migration and migration of FLSs. In vivo results corroborated this finding, with MAGL inhibition found to modulate pain and disease progression in an OA rat model. Collectively, these results indicate that MAGL administration is an ideal therapy treating OA.
Collapse
Affiliation(s)
- Xueyan Li
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, China
| | - Jing Zhou
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, 226000, China
| | - Liyuan Zhang
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Yi Shi
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Chun Zhang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 999, Xiwang Road, Shanghai, China
| | - Wen Sun
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China
| | - Miao Chu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, China
| | - Kai Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, China
| | - Chengyong Gu
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, China.
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, 226000, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou, Jiangsu, China.
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, 242, Guangji Road, Suzhou, Jiangsu, 226000, China.
| |
Collapse
|
63
|
Liu L, Tang H, Wang Y. Polymeric biomaterials: Advanced drug delivery systems in osteoarthritis treatment. Heliyon 2023; 9:e21544. [PMID: 38034809 PMCID: PMC10682535 DOI: 10.1016/j.heliyon.2023.e21544] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Polymeric biomaterials have emerged as a highly promising candidate for drug delivery systems (DDS), exhibiting significant potential to enhance the therapeutic landscape of osteoarthritis (OA) therapy. Their remarkable capacity to manifest desirable physicochemical attributes, coupled with their excellent biocompatibility and biodegradability, has greatly expanded their utility in pharmacotherapeutic applications. Nevertheless, an urgent necessity exists for a comprehensive synthesis of the most recent advances in polymeric DDS, providing valuable guidance for their implementation in the context of OA therapy. This review is dedicated to summarizing and examining recent developments in the utilization of polymeric DDS for OA therapy. Initially, we present an overview of the intricate pathophysiology characterizing OA and underscore the prevailing limitations inherent to current treatment modalities. Subsequently, we introduce diverse categories of polymeric DDS, including hydrogels, nanofibers, and microspheres, elucidating their inherent advantages and limitations. Moreover, we discuss and summarize the delivery of bioactive agents through polymeric biomaterials for OA therapy, emphasizing key findings and emerging trends. Finally, we highlight prospective directions for advancing polymeric DDS, offering a promising approach to enhance their translational potential for OA therapy.
Collapse
Affiliation(s)
- Lin Liu
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| | - Haifeng Tang
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| | - Yanjun Wang
- Department of Emergency, Honghui Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
| |
Collapse
|
64
|
Zhao H, Lin X, Chen Q, Wang X, Wu Y, Zhao X. Quercetin inhibits the NOX2/ROS-mediated NF-κB/TXNIP signaling pathway to ameliorate pyroptosis of cardiomyocytes to relieve sepsis-induced cardiomyopathy. Toxicol Appl Pharmacol 2023; 477:116672. [PMID: 37648089 DOI: 10.1016/j.taap.2023.116672] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/18/2023] [Accepted: 08/27/2023] [Indexed: 09/01/2023]
Abstract
Sepsis-induced cardiomyopathy (SIC) has high morbidity and mortality. Quercetin (QUE) has been used to treat many inflammatory diseases related to pyroptosis. However, its effect on SIC has not been reported before. We aimed to explore the therapeutic mechanism of QUE on SIC. We found that the expression levels of NOX2, markers of myocardial injury and inflammatory factors related to pyroptosis were upregulated in the serum of SIC patients. QUE improved the viability and reduced the death rate of LPS-treated H9C2 cells. It could downregulate the expression level of NOX2 and alleviate NOX2-induced mitochondrial damage to inhibit the ROS-mediated NF-κB/TXNIP pathway thus ameliorating cell pyroptosis. Overexpression of NOX2 partially attenuated the anti-pyroptotic effects of QUE on LPS-treated H9C2 cells in vitro. Besides, the results of animal experiments reported that the mitochondrial damage was reduced by QUE treatment, which subsequently inhibited the ROS-mediated NF-κB/TXNIP pathway to ameliorate cell pyroptosis to further alleviate myocardial injury in CLP-induced rats in vivo. To conclude, QUE suppressed the NOX2/ROS-mediated NF-κB/TXNIP signaling pathway to ameliorate pyroptosis of cardiomyocytes to relieve SIC.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhuaxi Road, Lixia District, Jinan City, Shandong Province 250014, China
| | - Xin Lin
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhuaxi Road, Lixia District, Jinan City, Shandong Province 250014, China
| | - Qingfeng Chen
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhuaxi Road, Lixia District, Jinan City, Shandong Province 250014, China
| | - Xiaoyue Wang
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhuaxi Road, Lixia District, Jinan City, Shandong Province 250014, China
| | - Yongya Wu
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhuaxi Road, Lixia District, Jinan City, Shandong Province 250014, China
| | - Xiaoxia Zhao
- Department of Emergency and Critical Care Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhuaxi Road, Lixia District, Jinan City, Shandong Province 250014, China.
| |
Collapse
|
65
|
Su J, Yu M, Wang H, Wei Y. Natural anti-inflammatory products for osteoarthritis: From molecular mechanism to drug delivery systems and clinical trials. Phytother Res 2023; 37:4321-4352. [PMID: 37641442 DOI: 10.1002/ptr.7935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 08/31/2023]
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects millions globally. The present nonsteroidal anti-inflammatory drug treatments have different side effects, leading researchers to focus on natural anti-inflammatory products (NAIPs). To review the effectiveness and mechanisms of NAIPs in the cellular microenvironment, examining their impact on OA cell phenotype and organelles levels. Additionally, we summarize relevant research on drug delivery systems and clinical randomized controlled trials (RCTs), to promote clinical studies and explore natural product delivery options. English-language articles were searched on PubMed using the search terms "natural products," "OA," and so forth. We categorized search results based on PubChem and excluded "natural products" which are mix of ingredients or compounds without the structure message. Then further review was separately conducted for molecular mechanisms, drug delivery systems, and RCTs later. At present, it cannot be considered that NAIPs can thoroughly prevent or cure OA. Further high-quality studies on the anti-inflammatory mechanism and drug delivery systems of NAIPs are needed, to determine the appropriate drug types and regimens for clinical application, and to explore the combined effects of different NAIPs to prevent and treat OA.
Collapse
Affiliation(s)
- Jianbang Su
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minghao Yu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Haochen Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yingliang Wei
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
66
|
Zou Z, Li H, Yu K, Ma K, Wang Q, Tang J, Liu G, Lim K, Hooper G, Woodfield T, Cui X, Zhang W, Tian K. The potential role of synovial cells in the progression and treatment of osteoarthritis. EXPLORATION (BEIJING, CHINA) 2023; 3:20220132. [PMID: 37933282 PMCID: PMC10582617 DOI: 10.1002/exp.20220132] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/15/2023] [Indexed: 11/08/2023]
Abstract
Osteoarthritis (OA), the commonest arthritis, is characterized by the progressive destruction of cartilage, leading to disability. The Current early clinical treatment strategy for OA often centers on anti-inflammatory or analgesia medication, weight loss, improved muscular function and articular cartilage repair. Although these treatments can relieve symptoms, OA tends to be progressive, and most patients require arthroplasty at the terminal stages of OA. Recent studies have shown a close correlation between joint pain, inflammation, cartilage destruction and synovial cells. Consequently, understanding the potential mechanisms associated with the action of synovial cells in OA could be beneficial for the clinical management of OA. Therefore, this review comprehensively describes the biological functions of synovial cells, the synovium, together with the pathological changes of synovial cells in OA, and the interaction between the cartilage and synovium, which is lacking in the present literature. Additionally, therapeutic approaches based on synovial cells for OA treatment are further discussed from a clinical perspective, highlighting a new direction in the treatment of OA.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Han Li
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
| | - Kai Yu
- Department of Bone and JointCentral Hospital of Zhuang He CityDalianLiaoningChina
| | - Ke Ma
- Department of Clinical MedicineChina Medical UniversityShenyangLiaoningChina
| | - Qiguang Wang
- National Engineering Research Center for BiomaterialsSichuan UniversityChengduSichuanChina
| | - Junnan Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Guozhen Liu
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
| | - Khoon Lim
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Gary Hooper
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Tim Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Xiaolin Cui
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- School of MedicineThe Chinese University of Hong Kong (Shenzhen)ShenzhenGuangdongChina
- Christchurch Regenerative Medicine and Tissue Engineering Group (CReaTE)Department of Orthopaedic Surgery and Musculoskeletal MedicineUniversity of OtagoChristchurchNew Zealand
| | - Weiguo Zhang
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| | - Kang Tian
- Department of Sports MedicineThe First Affiliated Hospital of Dalian Medical UniversityDalianLiaoningChina
- Key Laboratory of Molecular Mechanisms for Repair and Remodeling of Orthopaedic DiseasesLiaoning ProvinceDalianLiaoningChina
| |
Collapse
|
67
|
Peng Y, Li Y, Yang Y, Shi T, Liu R, Luan Y, Yin C. The Role and Potential Regulatory Mechanism of STING Modulated Macrophage Apoptosis and Differentiation in Severe Acute Pancreatitis-Associated Lung Injury. J Interferon Cytokine Res 2023; 43:455-468. [PMID: 37819622 DOI: 10.1089/jir.2023.0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
This study aims to investigate the role of STING in promoting macrophage apoptosis and regulating macrophage polarization in severe acute pancreatitis (SAP)-associated lung injury in vitro and in vivo. A murine model was established by intraperitoneal injection of caerulein and lipopolysaccharide (LPS). Meanwhile, ANA-1 cells were stimulated with LPS to induce apoptosis in vitro. More primary alveolar macrophages underwent apoptosis and M1 macrophage polarization in the SAP group compared with the control group, which was reversed by inhibiting STING. When ANA-1 cells were induced into M2-type macrophages, the reduction of M1 macrophage markers was accompanied by a decrease of LPS-induced apoptosis. Finally, the inhibitory effect of C-176 on STING ameliorates lung injury and inflammation by adjusting macrophage polarization and rescuing apoptosis. Therefore, inhibiting STING could be a new therapeutic strategy for treating acute pancreatitis-associated lung injury.
Collapse
Affiliation(s)
- Yiqiu Peng
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yingying Li
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yuxi Yang
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Tingjuan Shi
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Ruixia Liu
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yingyi Luan
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chenghong Yin
- Department of Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
68
|
Yamaura K, Nelson AL, Nishimura H, Rutledge JC, Ravuri SK, Bahney C, Philippon MJ, Huard J. Therapeutic potential of senolytic agent quercetin in osteoarthritis: A systematic review and meta-analysis of preclinical studies. Ageing Res Rev 2023; 90:101989. [PMID: 37442369 DOI: 10.1016/j.arr.2023.101989] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Quercetin, a natural flavonoid, has shown promise as a senolytic agent for various degenerative diseases. Recently, its protective effect against osteoarthritis (OA), a representative age-related disease of the musculoskeletal system, has attracted much attention. The aim of this study is to summarize and analyze the current literature on the effects of quercetin on OA cartilage in in vivo preclinical studies. METHODS The Medline (via/using PubMed), Embase, and Web of Science databases were searched up to March 10th, 2023. Risk of bias and the qualitative assessment including mechanisms of all eligible studies and a meta-analysis of cartilage histological scores among the applicable studies was performed. RESULTS A total of 12 in vivo animal studies were included in this systematic review. A random-effects meta-analysis was performed on six studies using the Osteoarthritis Research Society International (OARSI) scoring system, revealing that quercetin significantly improved OA cartilage OARSI scores (SMD, -6.30 [95% CI, -9.59 to -3.01]; P = 0.0002; heterogeneity: I2 = 86%). The remaining six studies all supported quercetin's protective effects against OA during disease and aging. CONCLUSIONS Quercetin has shown beneficial effects on cartilage during OA across animal species. Future double-blind randomized controlled clinical trials are needed to verify the efficacy of quercetin in the treatment of OA in humans.
Collapse
Affiliation(s)
- Kohei Yamaura
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA; Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Anna Laura Nelson
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| | - Haruki Nishimura
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA; Department of Orthopaedic Surgery, University Hospital of Occupational and Environmental Health, Fukuoka, Japan
| | - Joan C Rutledge
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| | - Sudheer K Ravuri
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA
| | - Chelsea Bahney
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA; The Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Marc J Philippon
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA; The Steadman Clinic, Vail, CO, USA
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO, USA.
| |
Collapse
|
69
|
Liu R, Zhou Y, Chen H, Xu H, Zuo M, Chen B, Wang H. Membrane vesicles from Lactobacillus johnsonii delay osteoarthritis progression via modulating macrophage glutamine synthetase/mTORC1 axis. Biomed Pharmacother 2023; 165:115204. [PMID: 37499456 DOI: 10.1016/j.biopha.2023.115204] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
AIMS The manipulation of macrophage recruitment and their shift in the M1/M2 ratio is a promising approach to mitigate osteoarthritis (OA). Nevertheless, the current clinical medication available for OA is only palliative and may result in undesirable outcomes. Hence, it is urgent to explore alternative disease-modifying drug supplement that are both safer and more effective in OA treatment, like probiotic and probiotic-derived membrane vesicles. METHODS The synovial inflammation and cartilage damage in collagenase-induced OA (CIOA) mice were observed using haematoxylin and eosin, saffron O-solid green and immunohistochemical staining. Bipedal balance test and open field test were conducted to determine the effectiveness of L. johnsonii-derived membrane vesicles (LJ-MVs) in reducing joint pain of CIOA mice. Additionally, Transwell, western blot, and immunological testing were used to examine the effect of LJ-MVs on macrophage migration and reprogramming. Furthermore, a 4D label-free proteomic analysis of LJ-MVs and their parent bacterium was performed, and the glutamine synthetase (GS)/mTORC1 axis in macrophage was verified by western blot. RESULTS L. johnsonii and its membrane vesicles, LJ-MVs, exhibit a novel ability to mitigate inflammation, cartilage damage, and pain associated with OA. This is achieved by their ability to impede macrophage migration, M1-like polarization, and inflammatory mediators secretion, while simultaneously promoting the M2/M1 ratio in synovial macrophages. The mechanism underlying this effect involves the modulation of macrophage GS/mTORC1 pathway, at least partially. SIGNIFICANCE Owing to their probiotic derivation, LJ-MVs will be a more dependable and potent disease-modifying drugs for the prevention and therapy of OA in the long run.
Collapse
Affiliation(s)
- Rangru Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Department of Spine Surgery of The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China; Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, China
| | - Yue Zhou
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Department of Spine Surgery of The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Huanxiong Chen
- Department of Spine Surgery, Hainan Province Clinical Medical Center, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Haixia Xu
- Department of Spine Surgery, Hainan Province Clinical Medical Center, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Min Zuo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Department of Spine Surgery of The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China
| | - Bo Chen
- Hainan Provincial Key Laboratory of R&D of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, China
| | - Hua Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine, Department of Spine Surgery of The First Affiliated Hospital, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
70
|
Luo P, Du M, Sun Q, Zhao T, He H. IL-38 suppresses macrophage M1 polarization to ameliorate synovial inflammation in the TMJ via GLUT-1 inhibition. Int Immunopharmacol 2023; 122:110619. [PMID: 37463548 DOI: 10.1016/j.intimp.2023.110619] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/25/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023]
Abstract
OBJECTIVES Interleukin (IL)-38 was discovered as an anti-inflammatory factor. However, IL-38's role in M1 macrophage polarization in the temporomandibular joint (TMJ) and the related mechanism are still unclear. We aimed to explore the effect and the mechanism of IL-38 on synovial inflammation in the TMJ in this study. METHODS The expression of IL-38 in the TMJ synovium and macrophages was determined using immunohistochemistry (IHC) and Western blotting (WB). M1 macrophage polarization was induced by LPS, the macrophages were pre-treated with IL-38, and the levels of inflammatory markers associated with M1 macrophages were measured. To assess the mechanism of IL-38, small-interfering RNA (siRNA)-GLUT-1 and STF31 were administered to macrophages, and the affected pathways were identified by WB. The effect of macrophage-conditioned medium (CM) on chondrocyte function was also determined. Finally, a mouse model of CFA-induced TMJ inflammation was established. Histological staining and IHC were used to determine the effect of IL-38. RESULTS IL-38 was detected at high levels in macrophages after lipopolysaccharide (LPS)challenge, and IL-38 downregulated M1 macrophage-related proinflammatory markers (iNOS, IL-6, TNF-α, and COX-2) in vitro. IL-38 suppressed M1 polarization by inhibiting GLUT-1 expression, NF-κB signaling, and MAPK signaling. Intriguingly, CM from macrophages that were pretreated with IL-38 and STF31 decreased inflammatory protein expression in chondrocytes. In addition, intra-articular injection of recombinant IL-38 ameliorated synovial inflammation in the TMJ by inhibiting M1 macrophage polarization and suppressing cartilage inflammation in vivo. CONCLUSIONS IL-38 is a novel anti-inflammatory factor that contributes to alleviating TMJ inflammation by inhibiting macrophage M1 polarization, thereby ameliorating chondrocyte inflammation and restoring TMJ homeostasis.
Collapse
Affiliation(s)
- Ping Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingyuan Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.; Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiao Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Tingting Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.; Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Hong He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.; Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
71
|
Tong X, Wang Y, Dong B, Li Y, Lang S, Ma J, Ma X. Effects of genus Epimedium in the treatment of osteoarthritis and relevant signaling pathways. Chin Med 2023; 18:92. [PMID: 37525296 PMCID: PMC10388486 DOI: 10.1186/s13020-023-00788-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/25/2023] [Indexed: 08/02/2023] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative joint disease in clinical practice with a high prevalence, especially in the elderly. Traditional Chinese Medicine (TCM) believes that OA belongs to the category of "Bi syndrome" and the "bone Bi syndrome". The etiology and pathogenesis lie in the deficiency of the liver and kidney, the deficiency of Qi and blood, and external exposure to wind, cold, and dampness. Epimedium is a yang-reinforcing herb in TCM, which can tonify the liver and kidney, strengthen muscles and bones, dispel wind, cold and dampness, and can treat both the symptoms and the root cause of "bone Bi syndrome". In addition, Epimedium contains a large number of ingredients. Through modern science and technology, more than 270 compounds have been found in Epimedium, among which flavonoids are the main active ingredients. Therefore, our study will review the effects and mechanisms of genus Epimedium in treating OA from two aspects: (1) Introduction of Epimedium and its main active ingredients; (2) Effects of Epimedium and its active ingredients in treating OA and relevant signaling pathways, in order to provide more ideas for OA treatment.
Collapse
Affiliation(s)
- Xue Tong
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yan Wang
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China
- Tianjin Hospital, Tianjin University, Tianjin, China
| | - Benchao Dong
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China
- Tianjin Hospital, Tianjin University, Tianjin, China
| | - Yan Li
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China
- Tianjin Hospital, Tianjin University, Tianjin, China
| | - Shuang Lang
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Hospital, Tianjin University, Tianjin, China
| | - Jianxiong Ma
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China.
- Tianjin Hospital, Tianjin University, Tianjin, China.
| | - Xinlong Ma
- Orthopaedics Institute of Tianjin, Tianjin Hospital, Tianjin, China.
- Tianjin Hospital, Tianjin University, Tianjin, China.
| |
Collapse
|
72
|
Yang L, Yu X, Liu M, Cao Y. A comprehensive analysis of biomarkers associated with synovitis and chondrocyte apoptosis in osteoarthritis. Front Immunol 2023; 14:1149686. [PMID: 37545537 PMCID: PMC10401591 DOI: 10.3389/fimmu.2023.1149686] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction Osteoarthritis (OA) is a chronic disease with high morbidity and disability rates whose molecular mechanism remains unclear. This study sought to identify OA markers associated with synovitis and cartilage apoptosis by bioinformatics analysis. Methods A total of five gene-expression profiles were selected from the Gene Expression Omnibus database. We combined the GEO with the GeneCards database and performed Gene Ontology and Kyoto Encyclopedia of Genes and Genome analyses; then, the least absolute shrinkage and selection operator (LASSO) algorithm was used to identify the characteristic genes, and a predictive risk score was established. We used the uniform manifold approximation and projection (UMAP) method to identify subtypes of OA patients, while the CytoHubba algorithm and GOSemSim R package were used to screen out hub genes. Next, an immunological assessment was performed using single-sample gene set enrichment analysis and CIBERSORTx. Results A total of 56OA-related differential genes were selected, and 10 characteristic genes were identified by the LASSO algorithm. OA samples were classified into cluster 1 and cluster 2 subtypes byUMAP, and the clustering results showed that the characteristic genes were significantly different between these groups. MYOC, CYP4B1, P2RY14, ADIPOQ, PLIN1, MFAP5, and LYVE1 were highly expressed in cluster 2, and ANKHLRC15, CEMIP, GPR88, CSN1S1, TAC1, and SPP1 were highly expressed in cluster 1. Protein-protein interaction network analysis showed that MMP9, COL1A, and IGF1 were high nodes, and the differential genes affected the IL-17 pathway and tumor necrosis factor pathway. The GOSemSim R package showed that ADIPOQ, COL1A, and SPP1 are closely related to the function of 31 hub genes. In addition, it was determined that mmp9 and Fos interact with multiple transcription factors, and the ssGSEA and CIBERSORTx algorithms revealed significant differences in immune infiltration between the two OA subtypes. Finally, a qPCR experiment was performed to explore the important genes in rat cartilage and synovium tissues; the qPCR results showed that COL1A and IL-17A were both highly expressed in synovitis tissues and cartilage tissues of OA rats, which is consistent with the predicted results. Discussion In the future, common therapeutic targets might be found forsimultaneous remissions of both phenotypes of OA.
Collapse
Affiliation(s)
- Ling Yang
- Department of Hematology, The First People’s Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Traditional Chinese Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyuan Yu
- Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Meng Liu
- Department of Clinical Laboratory,The First Affiliated Hospital of Xi’an Jiao Tong University, Xi’an, China
| | - Yang Cao
- Department of Hematology, The First People’s Hospital of Changzhou, Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
73
|
Suzuki T, Ohishi T, Tanabe H, Miyoshi N, Nakamura Y. Anti-Inflammatory Effects of Dietary Polyphenols through Inhibitory Activity against Metalloproteinases. Molecules 2023; 28:5426. [PMID: 37513300 PMCID: PMC10385587 DOI: 10.3390/molecules28145426] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent metalloproteinases that play important roles in a variety of diseases, including cancer, cardiovascular disease, diabetes, obesity, and brain diseases. Dietary polyphenols are thought to have a variety of beneficial effects on these diseases characterized by inflammation. Clinical studies have demonstrated that MMPs are in most cases upregulated in various inflammatory diseases, including osteoarthritis, rheumatoid arthritis, inflammatory bowel disease, and Alzheimer's disease. Studies using patient-derived human samples, animal studies, and cellular experiments have suggested that polyphenols may be beneficial against inflammatory diseases by suppressing MMP gene expression and enzyme activity. One important mechanism by which polyphenols exert their activity is the downregulation of reactive oxygen species that promote MMP expression. Another important mechanism is the direct binding of polyphenols to MMPs and their inhibition of enzyme activity. Molecular docking analyses have provided a structural basis for the interaction between polyphenols and MMPs and will help to explore new polyphenol-based drugs with anti-inflammatory properties.
Collapse
Affiliation(s)
- Takuji Suzuki
- Department of Food Science and Nutrition, Faculty of Human Life and Science, Doshisha Women's College of Liberal Arts, Kamigyo-ku, Kyoto 602-0893, Japan
| | - Tomokazu Ohishi
- Laboratory of Oncology, Institute of Microbial Chemistry (BIKAKEN), Microbial Chemistry Research Foundation, Shinagawa, Tokyo 141-0021, Japan
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu, Shizuoka 410-0301, Japan
| | - Hiroki Tanabe
- Department of Nutritional Sciences, Faculty of Health and Welfare Science, Nayoro City University, Nayoro, Hokkaido 096-8641, Japan
| | - Noriyuki Miyoshi
- Graduate School of Integrated Pharmaceutical and Nutritional Sciences, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoriyuki Nakamura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
74
|
Muthu S, Korpershoek JV, Novais EJ, Tawy GF, Hollander AP, Martin I. Failure of cartilage regeneration: emerging hypotheses and related therapeutic strategies. Nat Rev Rheumatol 2023; 19:403-416. [PMID: 37296196 DOI: 10.1038/s41584-023-00979-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/12/2023]
Abstract
Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.
Collapse
Affiliation(s)
- Sathish Muthu
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, New Delhi, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Jasmijn V Korpershoek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Emanuel J Novais
- Unidade Local de Saúde do Litoral Alentejano, Orthopedic Department, Santiago do Cacém, Portugal
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gwenllian F Tawy
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, UK
| | - Anthony P Hollander
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
75
|
Fang W, Yang M, Liu M, Jin Y, Wang Y, Yang R, Wang Y, Zhang K, Fu Q. Review on Additives in Hydrogels for 3D Bioprinting of Regenerative Medicine: From Mechanism to Methodology. Pharmaceutics 2023; 15:1700. [PMID: 37376148 PMCID: PMC10302687 DOI: 10.3390/pharmaceutics15061700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The regeneration of biological tissues in medicine is challenging, and 3D bioprinting offers an innovative way to create functional multicellular tissues. One common way in bioprinting is bioink, which is one type of the cell-loaded hydrogel. For clinical application, however, the bioprinting still suffers from satisfactory performance, e.g., in vascularization, effective antibacterial, immunomodulation, and regulation of collagen deposition. Many studies incorporated different bioactive materials into the 3D-printed scaffolds to optimize the bioprinting. Here, we reviewed a variety of additives added to the 3D bioprinting hydrogel. The underlying mechanisms and methodology for biological regeneration are important and will provide a useful basis for future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kaile Zhang
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| | - Qiang Fu
- Department of Urology, Affiliated Sixth People’s Hospital, Shanghai Jiaotong University, No. 600 Yi-Shan Road, Shanghai 200233, China; (W.F.); (M.Y.)
| |
Collapse
|
76
|
Chen Z, Jiang Y, Wu S, Dang M. Comprehensive analysis of femoral head necrosis based on machine learning and bioinformatics analysis. Medicine (Baltimore) 2023; 102:e33963. [PMID: 37335681 DOI: 10.1097/md.0000000000033963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/21/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a kind of disabling disease, given that the molecular mechanism of ONFH has not been elucidated, it is of significance to use bioinformatics analysis to understand the disease mechanism of ONFH and discover biomarkers. Gene set for ONFH GSE74089 was downloaded in the Gene Expression Omnibus, and "limma" package in R software was used to identify differentially expressed genes related to oxidative stress. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyze were performed for functional analysis. We constructed a protein interaction network and identified potential transcription factors and therapeutic drugs for the hub genes, and delineated the TF-hub genes network. Least absolute shrinkage and selection operator regression, support vector machine and cytoHubba were used to screen feature genes and key genes, which were validated by Receiver operating characteristic. CIBERSORT was used to explored the immune microenvironment. Subsequently, we identified the function of key genes using Gene set variation analysis and their relationship with each type of immune cell. Finally, molecular docking validated the binding association between molecules and validated genes. We detected 144 differentially expressed oxidative stress-related genes, and enrichment analysis showed that they were enriched in reactive oxygen species and AGE-RAGE signaling pathway. Protein-protein interaction and TF-hub genes network were conducted. Further exploration suggested that APOD and TMEM161A were feature genes, while TNF, NOS3 and CASP3 were key genes. Receiver operating characteristic analysis showed that APOD, CASP3, NOS3, and TNF have strong diagnostic ability. The key genes were enriched in oxidative phosphorylation. CIBERSORT analysis showed that 17 types immune cells were differentially relocated, and most of which were also closely related to key genes. In addition, genistein maybe potential therapeutic compound. In all, we identified that TNF, NOS3, and CASP3 played key roles on ONFH, and APOD, CASP3, NOS3, and TNF could serve as diagnostic biomarkers.
Collapse
Affiliation(s)
- Zheng Chen
- Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, China
| | - Yuankang Jiang
- Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, China
| | - Suwen Wu
- Guangzhou University of Chinese Medicine Third Clinical Medical College, Guangzhou, China
| | - Meng Dang
- Department of Anesthesiology, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
77
|
Ma J, Zhao W, Pei X, Li X, Zhao W. MicroRNA-345-3p is a potential biomarker and ameliorates rheumatoid arthritis by reducing the release of proinflammatory cytokines. J Orthop Surg Res 2023; 18:399. [PMID: 37264454 DOI: 10.1186/s13018-023-03797-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/13/2023] [Indexed: 06/03/2023] Open
Abstract
OBJECTIVES The study was to explore the influence of microRNA (miR)-345-3p on proinflammatory cytokines in patients with rheumatoid arthritis (RA). METHODS A total of 32 RA patients and 32 healthy patients were enrolled. Proinflammatory factors in patients' serum were detected by ELISA, and miR-345-3p was detected by RT-qPCR. The correlation between miR-345-3p expression and proinflammatory factors in RA patients was analyzed. The diagnostic value of miR-345-3p and proinflammatory factors in RA patients was analyzed by receiver operating curve diagnosis. The predictive value of miR-345-3p levels and proinflammatory factors in RA patients was analyzed by multivariate Cox regression. HFLS-RA and HFLS cells were cultured, in which miR-345-3p and proinflammatory cytokines were detected by RT-qPCR. Cell proliferation and apoptosis were determined by CCK-8 and flow cytometry, respectively. RESULTS MiR-345-3p was lowly expressed in the serum of RA patients. MiR-345-3p and proinflammatory factors were of diagnostic and predictive values in RA. Elevated miR-345-3p restrained the production of proinflammatory factors of HFLS-RA cells, improved cell proliferation, and reduced apoptosis. CONCLUSION MiR-345-3p is a potential biomarker and ameliorates RA by reducing the release of proinflammatory cytokines.
Collapse
Affiliation(s)
- Jun Ma
- Department of Orthopedics, Jiu Quan People's Hospital, No. 22, West Street, Suzhou District, Jiu Quan City, 735000, Gansu Province, China
| | - Wei Zhao
- Department of Orthopedics, Jiu Quan People's Hospital, No. 22, West Street, Suzhou District, Jiu Quan City, 735000, Gansu Province, China
| | - Xue Pei
- Department of Orthopedics, Jiu Quan People's Hospital, No. 22, West Street, Suzhou District, Jiu Quan City, 735000, Gansu Province, China
| | - XinZhi Li
- Department of Orthopaedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang City, 443001, Hubei Province, China
| | - Wei Zhao
- Department of Orthopedics, Jiu Quan People's Hospital, No. 22, West Street, Suzhou District, Jiu Quan City, 735000, Gansu Province, China.
| |
Collapse
|
78
|
Xiong B, Chen L, Huang Y, Lu G, Chen C, Nong J, Pan H. ZBTB16 eases lipopolysaccharide‑elicited inflammation, apoptosis and degradation of extracellular matrix in chondrocytes during osteoarthritis by suppressing GRK2 transcription. Exp Ther Med 2023; 25:276. [PMID: 37206562 PMCID: PMC10189728 DOI: 10.3892/etm.2023.11975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/23/2023] [Indexed: 05/21/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease of the bone that is a major contributor of disability in the elderly population. Zinc finger and BTB domain-containing 16 (ZBTB16) is a transcription factor that has been previously revealed to be impaired in human OA tissues. The present study was designed to elaborate the potential impact of ZBTB16 on OA and to possibly assess any latent regulatory mechanism. ZBTB16 expression in human OA tissues was examined using the Gene Expression Series (GSE) database (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE169077) whereas ZBTB16 expression in chondrocytes was examined using reverse transcription-quantitative PCR (RT-qPCR) and western blotting. Cell viability was examined using a Cell Counting Kit-8 assay. A TUNEL assay and western blotting were used to assess cell apoptosis and apoptosis-related markers, including Bcl-2, Bax and cleaved caspase-3. The levels and expression of inflammatory factors, including TNF-α, IL-1β and IL-6, were determined by ELISA and western blotting. RT-qPCR and western blotting were also used to analyze the expression levels of extracellular matrix (ECM)-degrading enzymes, including MMP-13, a disintegrin-like and metalloproteinase with thrombospondin type-1 motifs-5, aggrecan and collagen type II α1. After the potential binding of ZBTB16 with the G protein coupled receptor kinase type 2 (GRK2) promoter was predicted using the Cistrome DB database, GRK2 expression was confirmed by RT-qPCR and western blotting. Chromatin immunoprecipitation and luciferase reporter assays were then used to determine the potential interaction between ZBTB16 and the GRK2 promoter. Following GRK2 overexpression in ZBTB16-overexpressing chondrocytes by co-transfection of GRK2 and ZBTB16 overexpression plasmids, the aforementioned functional experiments were performed again. ZBTB16 expression was found to be reduced in human OA tissues compared with in normal cartilage tissues and lipopolysaccharide (LPS)-stimulated chondrocytes. ZBTB16 overexpression increased cell viability whilst decreasing apoptosis, inflammation and ECM degradation by LPS-treated chondrocytes. In addition, GRK2 expression was found to be increased in LPS-stimulated chondrocytes. ZBTB16 successfully bound to the GRK2 promoter, which negatively modulated GRK2 expression. GRK2 upregulation reversed the effects of ZBTB16 overexpression on the viability, apoptosis, inflammation and ECM degradation by LPS-challenged chondrocytes. In conclusion, these data suggest that ZBTB16 may inhibit the development of OA through the transcriptional inactivation of GRK2.
Collapse
Affiliation(s)
- Bo Xiong
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Lihua Chen
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Yue Huang
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Guanyu Lu
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Cai Chen
- Department of Orthopaedics and Traumatology, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Jiao Nong
- Teaching Department, First Affiliated Hospital of The Guangxi University of Chinese Medicine, Nanning, Guangxi 530000, P.R. China
| | - Haida Pan
- Department of Orthopaedics and Traumatology, Yongjia County Traditional Chinese Medicine Hospital, Wenzhou, Zhejiang 325100, P.R. China
- Correspondence to: Dr Haida Pan, Department of Orthopaedics and Traumatology, Yongjia County Traditional Chinese Medicine Hospital, 6 Park Road, Jiangbei Street, Yongjia, Wenzhou, Zhejiang 325100, P.R. China
| |
Collapse
|
79
|
Yang XL, Wang CX, Wang JX, Wu SM, Yong Q, Li K, Yang JR. In silico evidence implicating novel mechanisms of Prunella vulgaris L . as a potential botanical drug against COVID-19-associated acute kidney injury. Front Pharmacol 2023; 14:1188086. [PMID: 37274117 PMCID: PMC10232756 DOI: 10.3389/fphar.2023.1188086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
COVID-19-associated acute kidney injury (COVID-19 AKI) is an independent risk factor for in-hospital mortality and has the potential to progress to chronic kidney disease. Prunella vulgaris L., a traditional Chinese herb that has been used for the treatment of a variety of kidney diseases for centuries, could have the potential to treat this complication. In this study, we studied the potential protective role of Prunella vulgaris in COVID-19 AKI and explored its specific mechanisms applied by network pharmacology and bioinformatics methods. The combination of the protein-protein interaction network and Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment -target gene network revealed eight key target genes (VEGFA, ICAM1, IL6, CXCL8, IL1B, CCL2, IL10 and RELA). Molecular docking showed that all these eight gene-encoded proteins could be effectively bound to three major active compounds (quercetin, luteolin and kaempferol), thus becoming potential therapeutic targets. Molecular dynamics simulation also supports the binding stability of RELA-encoded protein with quercetin and luteolin. Together, our data suggest that IL6, VEGFA, and RELA could be the potential drug targets by inhibiting the NF-κB signaling pathway. Our in silico studies shed new insights into P. vulgaris and its ingredients, e.g., quercetin, as potential botanical drugs against COVID-19 AKI, and warrant further studies on efficacy and mechanisms.
Collapse
Affiliation(s)
- Xue-Ling Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-Xuan Wang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jia-Xing Wang
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shi-Min Wu
- Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Qing Yong
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ju-Rong Yang
- Department of Nephrology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
80
|
Sun Z, Xiong H, Lou T, Liu W, Xu Y, Yu S, Wang H, Liu W, Yang L, Zhou C, Fan C. Multifunctional Extracellular Matrix Hydrogel with Self-Healing Properties and Promoting Angiogenesis as an Immunoregulation Platform for Diabetic Wound Healing. Gels 2023; 9:gels9050381. [PMID: 37232972 DOI: 10.3390/gels9050381] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
Treating chronic wounds is a global challenge. In diabetes mellitus cases, long-time and excess inflammatory responses at the injury site may delay the healing of intractable wounds. Macrophage polarization (M1/M2 types) can be closely associated with inflammatory factor generation during wound healing. Quercetin (QCT) is an efficient agent against oxidation and fibrosis that promotes wound healing. It can also inhibit inflammatory responses by regulating M1-to-M2 macrophage polarization. However, its limited solubility, low bioavailability, and hydrophobicity are the main issues restricting its applicability in wound healing. The small intestinal submucosa (SIS) has also been widely studied for treating acute/chronic wounds. It is also being extensively researched as a suitable carrier for tissue regeneration. As an extracellular matrix, SIS can support angiogenesis, cell migration, and proliferation, offering growth factors involved in tissue formation signaling and assisting wound healing. We developed a series of promising biosafe novel diabetic wound repair hydrogel wound dressings with several effects, including self-healing properties, water absorption, and immunomodulatory effects. A full-thickness wound diabetic rat model was constructed for in vivo assessment of QCT@SIS hydrogel, in which hydrogels achieved a markedly increased wound repair rate. Their effect was determined by the promotion of the wound healing process, the thickness of granulation tissue, vascularization, and macrophage polarization during wound healing. At the same time, we injected the hydrogel subcutaneously into healthy rats to perform histological analyses of sections of the heart, spleen, liver, kidney, and lung. We then tested the biochemical index levels in serum to determine the biological safety of the QCT@SIS hydrogel. In this study, the developed SIS showed convergence of biological, mechanical, and wound-healing capabilities. Here, we focused on constructing a self-healing, water-absorbable, immunomodulatory, and biocompatible hydrogel as a synergistic treatment paradigm for diabetic wounds by gelling the SIS and loading QCT for slow drug release.
Collapse
Affiliation(s)
- Zhenghua Sun
- Graduate School, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Hao Xiong
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Tengfei Lou
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Weixuan Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Yi Xu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Shiyang Yu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Hui Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Wanjun Liu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
| | - Liang Yang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Chao Zhou
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| | - Cunyi Fan
- Graduate School, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, China
- Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Building 3, Langu Science and Technology Park, Lane 70, Haiji 6th Road, Shanghai 201306, China
| |
Collapse
|
81
|
Wei H, Qin J, Huang Q, Jin Z, Zheng L, Zhao J, Qin Z. Epigallocatechin-3-gallate (EGCG) based metal-polyphenol nanoformulations alleviates chondrocytes inflammation by modulating synovial macrophages polarization. Biomed Pharmacother 2023; 161:114366. [PMID: 36857913 DOI: 10.1016/j.biopha.2023.114366] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
The activation of M1-type macrophages are dominant cells secreting proinflammatory present within the inflamed synovium in the progression of osteoarthritis (OA). Increased oxidative stress, such as redundant ROS and hydrogen peroxide (H2O2), are important factors in driving macrophages to polarize into M1 type. In this study, metal-polyphenol nanoformulations (Cu-Epigallocatechin-3-gallate (Cu-EGCG) nanosheets) were synthesized through the coordination interaction between EGCG and copper ions, which possessed the antioxidant effect of EGCG and anti-inflammatory of Cu2+. Results showed that Cu-EGCG nanosheets were biocompatible and the Cu2+ could be sustained released from the nanoparticles. Cu-EGCG nanosheets with multienzyme-like antioxidative activity could effectively scavenge the excessive intracellular ROS, leading to significantly decreased expression of the pro-inflammatory cytokines, which could reduce the expression of M1-type macrophages and exhibit excellent promotion on shifting macrophages to M2 phenotypes. Moreover, the secreted factor from the cell supernatant of Cu-EGCG treated macrophages exhibited anti-inflammatory potential in chondrocytes of inflamed synovial joints. This study suggests a novel strategy for OA therapy by using metal-polyphenol nanoformulations targeting macrophages.
Collapse
Affiliation(s)
- Hong Wei
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jun Qin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Quanxin Huang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Zhiqiang Jin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China; Research Centre for Regenerative Medicine, Department of Orthopedics, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China.
| | - Zainen Qin
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China.
| |
Collapse
|
82
|
Wang H, Su J, Yu M, Xia Y, Wei Y. PGC-1α in osteoarthritic chondrocytes: From mechanism to target of action. Front Pharmacol 2023; 14:1169019. [PMID: 37089944 PMCID: PMC10117990 DOI: 10.3389/fphar.2023.1169019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023] Open
Abstract
Osteoarthritis (OA) is one of the most common degenerative joint diseases, often involving the entire joint. The degeneration of articular cartilage is an important feature of OA, and there is growing evidence that the mitochondrial biogenesis master regulator peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) exert a chondroprotective effect. PGC-1α delays the development and progression of OA by affecting mitochondrial biogenesis, oxidative stress, mitophagy and mitochondrial DNA (mtDNA) replication in chondrocytes. In addition, PGC-1α can regulate the metabolic abnormalities of OA chondrocytes and inhibit chondrocyte apoptosis. In this paper, we review the regulatory mechanisms of PGC-1α and its effects on OA chondrocytes, and introduce potential drugs and novel nanohybrid for the treatment of OA which act by affecting the activity of PGC-1α. This information will help to further elucidate the pathogenesis of OA and provide new ideas for the development of therapeutic strategies for OA.
Collapse
Affiliation(s)
- Haochen Wang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianbang Su
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minghao Yu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Xia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yang Xia, ; Yingliang Wei,
| | - Yingliang Wei
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yang Xia, ; Yingliang Wei,
| |
Collapse
|
83
|
Yan Y, Lu A, Dou Y, Zhang Z, Wang X, Zhai L, Ai L, Du M, Jiang L, Zhu Y, Shi Y, Liu X, Jiang D, Wang J. Nanomedicines Reprogram Synovial Macrophages by Scavenging Nitric Oxide and Silencing CA9 in Progressive Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207490. [PMID: 36748885 PMCID: PMC10104675 DOI: 10.1002/advs.202207490] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/12/2023] [Indexed: 06/18/2023]
Abstract
Osteoarthritis (OA) is a progressive joint disease characterized by inflammation and cartilage destruction, and its progression is closely related to imbalances in the M1/M2 synovial macrophages. A two-pronged strategy for the regulation of intracellular/extracellular nitric oxide (NO) and hydrogen protons for reprogramming M1/M2 synovial macrophages is proposed. The combination of carbonic anhydrase IX (CA9) siRNA and NO scavenger in "two-in-one" nanocarriers (NAHA-CaP/siRNA nanoparticles) is developed for progressive OA therapy by scavenging NO and inhibiting CA9 expression in synovial macrophages. In vitro experiments demonstrate that these NPs can significantly scavenge intracellular NO similar to the levels as those in the normal group and downregulate the expression levels of CA9 mRNA (≈90%), thereby repolarizing the M1 macrophages into the M2 phenotype and increasing the expression levels of pro-chondrogenic TGF-β1 mRNA (≈1.3-fold), and inhibiting chondrocyte apoptosis. Furthermore, in vivo experiments show that the NPs have great anti-inflammation, cartilage protection and repair effects, thereby effectively alleviating OA progression in both monoiodoacetic acid-induced early and late OA mouse models and a surgical destabilization of medial meniscus-induced OA rat model. Therefore, the siCA9 and NO scavenger "two-in-one" delivery system is a potential and efficient strategy for progressive OA treatment.
Collapse
Affiliation(s)
- Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yun Dou
- Department of Sports MedicinePeking University Third HospitalBeijing100191China
| | - Zhen Zhang
- Department of Sports MedicinePeking University Third HospitalBeijing100191China
| | - Xiang‐Yu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Lin Zhai
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Li‐Ya Ai
- Department of Sports MedicinePeking University Third HospitalBeijing100191China
| | - Ming‐Ze Du
- Department of Sports MedicinePeking University Third HospitalBeijing100191China
| | - Lin‐Xia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yuan‐Jun Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Yu‐Jie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Xiao‐Yan Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Dong Jiang
- Department of Sports MedicinePeking University Third HospitalBeijing100191China
| | - Jian‐Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemsState Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Laboratory of Innovative Formulations and Pharmaceutical ExcipientsNingbo Institute of Marine MedicinePeking UniversityBeijing315832China
| |
Collapse
|
84
|
Chen H, Liu J, Peng S, Yang G, Cheng X, Chen L, Zhang H, Zhao Y, Yao P, Tang Y. Autophagy and exosomes coordinately mediate quercetin's protective effects on alcoholic liver disease. J Nutr Biochem 2023; 116:109332. [PMID: 36965782 DOI: 10.1016/j.jnutbio.2023.109332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 12/09/2022] [Accepted: 03/18/2023] [Indexed: 03/27/2023]
Abstract
Alcoholic liver disease (ALD), a spectrum of liver abnormalities induced by chronic alcohol abuse, continues to be the major cause of life-threatening liver disease in developed countries. Autophagy and exosomes were individually confirmed to be involved in the pathogenesis of ALD. Here, we sought to identify the role of autophagy and exosomes in the liver protective effects of quercetin. We observed decreased hepatic LC3II/LC3I and increased p62 level in ethanol-fed mice, and these changes were alleviated by quercetin. Meanwhile, nanoparticle tracking analysis (NTA) showed elevated serum exosomes numbers in ethanol-fed mice, which was combated by quercetin. Ethanol induced elevated LDH, ALT, and AST in HepG2 supernatant, which was alleviated by cytochalasin D (exosomes uptake inhibitor). Moreover, quercetin reduced ethanol-induced LDH and ALT elevation in vitro, and the effects of quercetin were reversed by Rab27a overexpression (induce exosomes release) or wortmannin treatment (autophagy inhibitor). Transcriptomic analysis supported that quercetin reversed the change of lysosome related genes disturbed by ethanol. Meanwhile, western blot analysis exhibited decreased hepatic expression of LAMP2 and ATPA6V1B2, and active Cathepsin B/Cathepsin B by quercetin treatment, indicating quercetin alleviated lysosome dysfunction in ethanol-fed mice. Baf A treatment or transfection of siTFEB offset quercetin's effects in ethanol-induced LDH and ALT elevation, exosomes release, and autophagy inhibition (LC3II/I and p62 accumulation). Taken together, quercetin coordinately activates autophagy and combats exosomes release by restoring lysosome function, and further mitigates ethanol-induced liver damage.
Collapse
Affiliation(s)
- Huimin Chen
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Jingjing Liu
- Henan Provincial Center for Disease Control and Prevention, Zhengzhou, 450000, Henan, China
| | - Shufen Peng
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Guang Yang
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xueer Cheng
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Li Chen
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Han Zhang
- Department of Preventive Medicine, School of Health Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Ying Zhao
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Ping Yao
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; State Key Laboratory of Environment Health (Incubation), Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Key Laboratory of Environment & Health, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430030, Hubei, China
| | - Yuhan Tang
- Hubei Key Laboratory of Food Nutrition and Safety, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; State Key Laboratory of Environment Health (Incubation), Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China; Key Laboratory of Environment & Health, Huazhong University of Science and Technology, Ministry of Education, Wuhan, 430030, Hubei, China.
| |
Collapse
|
85
|
Preparation of inhalable quercetin-β-cyclodextrin inclusion complexes using the supercritical antisolvent process for the prevention of smoke inhalation-induced acute lung injury. J CO2 UTIL 2023. [DOI: 10.1016/j.jcou.2023.102414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
86
|
Yang T, Hu Y, Jiang W, Pang J, Zhou Y, Zhang H, Yin Z, Jiang Z, Qian S, Wei C, Yan M, Zhu X, Wang T, Lu Q. YY1 was indispensable for the alleviation of quercetin on diabetic nephropathy-associated tubulointerstitial inflammation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154659. [PMID: 36641979 DOI: 10.1016/j.phymed.2023.154659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND The emergence of tubulointerstitial inflammation (TI) could accelerate the development of tubulointerstitial fibrosis (TIF) of diabetic nephropathy (DN). Yin Yang 1 (YY1) was a new pro-inflammatory mediator and became the important target of DN-related TIF. Quercetin performed an effective role in anti-inflammation and was probable to bind to YY1. However, the role of YY1 in quercetin's anti-inflammatory effect on DN-related TIF was uncovered. PURPOSE To investigate the potential effect and mechanism of quercetin against DN-related TI. STUDY DESIGN AND METHODS The protein levels of YY1 were examined in the renal tubular epithelial cells (RTECs) of db/db mice and HG-cultured HK-2 cells. Molecular modeling studies and YY1 overexpression lentivirus vector were selected to further confirm the indispensable part of YY1 in quercetin's TI protection in vitro. Luciferase assay and chromatin immunoprecipitation (ChIP) assay were carried out to identify whether YY1 directly regulated IL-6/STAT3 signaling by binding to the IL-6 promoter in quercetin's TI protection in vitro. At last, the important role of YY1-mediated IL-6/STAT3 signaling in quercetin's TIF protection effect was further identified by using of YY1 overexpression lentivirus vector and IL-6 specific inhibitor tocilizumab. RESULTS Along with the alleviated tubulointerstitial injury by quercetin in the RTECs of db/db mice and HK-2 cells stimulated by HG, YY1-mediated IL-6/STAT-3 pathway involved in TI protection of quercetin in vivo and in vitro. Quercetin bound to YY1 and decreased its protein expression, and YY1 directly suppressed IL-6 transcription by bounding to its promoter, resulting in the alleviation of inflammation by inactivating of IL-6/STAT-3 pathway in vitro. YY1-mediated IL-6/STAT-3 pathway was also indispensable for the alleviation of quercetin on DN-associated TIF. CONCLUSION YY1 could not be absent from quercetin's anti-inflammatory effect on DN-associated TIF via alleviating IL-6/STAT-3 pathway mediated TI.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yinlu Hu
- Department of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi 214000, China
| | - Wenjie Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Jiale Pang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yequan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Huanming Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Zeyuan Yin
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, United Kingdom
| | - Zhenzhou Jiang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Sitong Qian
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Chujing Wei
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xia Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Tao Wang
- Department of Pharmacy, The affiliated hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
87
|
Zhang G, Qi C, Rong L, Ju C. Intra-articular delivery of antioxidative polymer-based nanospheres reduces intracellular reactive oxygen species in macrophages and alleviates cartilage damage in a rat model. J Biomater Appl 2023; 37:1384-1392. [PMID: 36342066 DOI: 10.1177/08853282221137774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cartilage damage is frequent in various joint diseases, mainly manifested by the loss of type II collagen and the degradation of proteoglycans. Diclofenac sodium is a commonly used drug for the treatment of joint diseases, but simple administration is often affected by drug clearance and rapid metabolism. Intra-articular drug delivery is an effective method for local enrichment of high concentration of drugs. However, due to the short half-life of diclofenac sodium, prolonging the stability and duration of the drug can alleviate the disadvantages of direct intra-articular application. Nanospheres for delivering drugs to treat joint diseases could be a remedy for cartilage damage. In addition, excessive production of reactive oxygen species (ROS) by macrophages activated in damaged cartilage would aggravate cartilage damage. Therefore, this study intends to use poly lactic-co-glycolic acid nanospheres to load and deliver diclofenac sodium to inhibit chondrocyte death while regulating the generation of ROS, thereby promoting the treatment of cartilage damage.
Collapse
Affiliation(s)
- Guihua Zhang
- Department of Bone and Joint Surgery, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| | - Chaopeng Qi
- Department of Hand and Microsurgery, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| | - Lingyan Rong
- Department of Pharmacy, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| | - Chuanbao Ju
- Department of Emergency Trauma, Wendeng Hospital of Traditional Chinese Orthopedics and Traumatology of Shandong Province, Shandong, China
| |
Collapse
|
88
|
Targeting macrophage polarization as a promising therapeutic strategy for the treatment of osteoarthritis. Int Immunopharmacol 2023; 116:109790. [PMID: 36736223 DOI: 10.1016/j.intimp.2023.109790] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) is a chronic osteoarthropathy characterized by the progressive degeneration of articular cartilage and synovial inflammation. Early OA clinical treatments involve intra-articular injection of glucocorticoids, oral acetaminophen and non-steroidal anti-inflammatory drugs (NSAIDs), which are used for anti-inflammation and pain relief. However, long-term use of these agents will lead to inevitable side effects, even aggravate cartilage loss. At present, there are no disease-modifying OA drugs (DMOADs) yet approved by regulatory agencies. Polarization regulation of synovial macrophages is a new target for OA treatment. Inhibiting M1 polarization and promoting M2 polarization of synovial macrophages can alleviate synovial inflammation, relieve joint pain and inhibit articular cartilage degradation, which is a promising strategy for OA treatment. In this study, we describe the molecular mechanisms of macrophage polarization and its key role in the development of OA. Subsequently, we summarize the latest progress of strategies for OA treatment through macrophage reprogramming, including small molecule compounds (conventional western medicine and synthetic compounds, monomer compounds of traditional Chinese medicine), biomacromolecules, metal/metal oxides, cells, and cell derivatives, and interprets the molecular mechanisms, hoping to provide some information for DMOADs development.
Collapse
|
89
|
Wang H, Yan Y, Pathak JL, Hong W, Zeng J, Qian D, Hao B, Li H, Gu J, Jaspers RT, Wu G, Shao M, Peng G, Lan H. Quercetin prevents osteoarthritis progression possibly via regulation of local and systemic inflammatory cascades. J Cell Mol Med 2023; 27:515-528. [PMID: 36722313 PMCID: PMC9930437 DOI: 10.1111/jcmm.17672] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 02/02/2023] Open
Abstract
Due to the lack of effective treatments, osteoarthritis (OA) remains a challenge for clinicians. Quercetin, a bioflavonoid, has shown potent anti-inflammatory effects. However, its effect on preventing OA progression and the underlying mechanisms are still unclear. In this study, Sprague-Dawley male rats were divided into five groups: control group, OA group (monosodium iodoacetate intra-articular injection), and three quercetin-treated groups. Quercetin-treated groups were treated with intragastric quercetin once a day for 28 days. Gross observation and histopathological analysis showed cartilage degradation and matrix loss in the OA group. High-dose quercetin-group joints showed failure in OA progression. High-dose quercetin inhibited the OA-induced expression of MMP-3, MMP-13, ADAMTS4, and ADAMTS5 and promoted the OA-reduced expression of aggrecan and collagen II. Levels of most inflammatory cytokines and growth factors tested in synovial fluid and serum were upregulated in the OA group and these increases were reversed by high-dose quercetin. Similarly, subchondral trabecular bone was degraded in the OA group and this effect was reversed in the high-dose quercetin group. Our findings indicate that quercetin has a protective effect against OA development and progression possibly via maintaining the inflammatory cascade homeostasis. Therefore, quercetin could be a potential therapeutic agent to prevent OA progression in risk groups.
Collapse
Affiliation(s)
- Haiyan Wang
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement SciencesVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Yongyong Yan
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement SciencesVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Janak L. Pathak
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina
| | - Wei Hong
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Jing Zeng
- Liwan Central Hospital of GuangzhouGuangzhouChina
| | - Dongyang Qian
- Department of Orthopaedics, The First Affiliated HospitalGuangzhou Medical University/Guangdong Key Laboratory of Orthopaedic Technology and Implant MaterialsGuangzhouChina
| | - Binwei Hao
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina,Department of Pulmonary and Critical Care Medicine, Shanxi Bethune HospitalShanxi Academy of Medical SciencesTaiyuanChina
| | - Haiqing Li
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jinlan Gu
- GMU‐GIBH Joint School of Life SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Richard T. Jaspers
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative MedicineGuangzhouChina,Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement SciencesVrije Universiteit Amsterdam, Amsterdam Movement SciencesAmsterdamThe Netherlands
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Centre for Dentistry Amsterdam (ACTA)Vrije Universiteit Amsterdam, Amsterdam Movement ScienceAmsterdamThe Netherlands
| | - Ming Shao
- Department of Orthopaedic SurgeryThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Gongyong Peng
- State Key Laboratory of Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Haifeng Lan
- Department of Orthopaedic SurgeryThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
90
|
Cao Z, Wang H, Chen J, Zhang Y, Mo Q, Zhang P, Wang M, Liu H, Bao X, Sun Y, Zhang W, Yao Q. Silk-based hydrogel incorporated with metal-organic framework nanozymes for enhanced osteochondral regeneration. Bioact Mater 2023; 20:221-242. [PMID: 35702612 PMCID: PMC9163388 DOI: 10.1016/j.bioactmat.2022.05.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Osteochondral defects (OCD) cannot be efficiently repaired due to the unique physical architecture and the pathological microenvironment including enhanced oxidative stress and inflammation. Conventional strategies, such as the control of implant microstructure or the introduction of growth factors, have limited functions failing to manage these complex environments. Here we developed a multifunctional silk-based hydrogel incorporated with metal-organic framework nanozymes (CuTA@SF) to provide a suitable microenvironment for enhanced OCD regeneration. The incorporation of CuTA nanozymes endowed the SF hydrogel with a uniform microstructure and elevated hydrophilicity. In vitro cultivation of mesenchymal stem cells (MSCs) and chondrocytes showed that CuTA@SF hydrogel accelerated cell proliferation and enhanced cell viability, as well as had antioxidant and antibacterial properties. Under the inflammatory environment with the stimulation of IL-1β, CuTA@SF hydrogel still possessed the potential to promote MSC osteogenesis and deposition of cartilage-specific extracellular matrix (ECM). The proteomics analysis further confirmed that CuTA@SF hydrogel promoted cell proliferation and ECM synthesis. In the full-thickness OCD model of rabbit, CuTA@SF hydrogel displayed successfully in situ OCD regeneration, as evidenced by micro-CT, histology (HE, S/O, and toluidine blue staining) and immunohistochemistry (Col I and aggrecan immunostaining). Therefore, CuTA@SF hydrogel is a promising biomaterial targeted at the regeneration of OCD.
Collapse
Affiliation(s)
- Zhicheng Cao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Hongmei Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Department of Pharmaceutical Sciences, Binzhou Medical University, 264003, Yantai, Shandong, China
| | - Jialin Chen
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Yanan Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Qingyun Mo
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Po Zhang
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Mingyue Wang
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Haoyang Liu
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Xueyang Bao
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Yuzhi Sun
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- School of Medicine, Southeast University, 210009, Nanjing, China
| | - Wei Zhang
- School of Medicine, Southeast University, 210009, Nanjing, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, 210096, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, 210006, Nanjing, China
- China Orthopedic Regenerative Medicine Group (CORMed), China
| |
Collapse
|
91
|
Renaudin F, Oudina K, Gerbaix M, McGilligan Subilia M, Paccaud J, Jaquet V, Krause KH, Ferrari S, Laumonier T, Hannouche D. NADPH oxidase 4 deficiency attenuates experimental osteoarthritis in mice. RMD Open 2023; 9:rmdopen-2022-002856. [PMID: 36810185 PMCID: PMC9945017 DOI: 10.1136/rmdopen-2022-002856] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/18/2023] [Indexed: 02/23/2023] Open
Abstract
OBJECTIVE Low-grade inflammation plays a pivotal role in osteoarthritis (OA) through exposure to reactive oxygen species (ROS). In chondrocytes, NADPH oxidase 4 (NOX4) is one of the major ROS producers. In this study, we evaluated the role of NOX4 on joint homoeostasis after destabilisation of the medial meniscus (DMM) in mice. METHODS Experimental OA was simulated on cartilage explants using interleukin-1β (IL-1β) and induced by DMM in wild-type (WT) and NOX4 knockout (NOX4-/-) mice. We evaluated NOX4 expression, inflammation, cartilage metabolism and oxidative stress by immunohistochemistry. Bone phenotype was also determined by micro-CT and histomorphometry. RESULTS Whole body NOX4 deletion attenuated experimental OA in mice, with a significant reduction of the OARSI score at 8 weeks. DMM increased total subchondral bone plate (SB.Th), epiphysial trabecular thicknesses (Tb.Th) and bone volume fraction (BV/TV) in both NOX4-/- and wild-type (WT) mice. Interestingly, DDM decreased total connectivity density (Conn.Dens) and increased medial BV/TV and Tb.Th only in WT mice. Ex vivo, NOX4 deficiency increased aggrecan (AGG) expression and decreased matrix metalloproteinase 13 (MMP13) and collagen type I (COL1) expression. IL-1β increased NOX4 and 8-hydroxy-2'-deoxyguanosine (8-OHdG) expression in WT cartilage explants but not in NOX4-/-. In vivo, absence of NOX4 increased anabolism and decreased catabolism after DMM. Finally, NOX4 deletion decreased synovitis score, 8-OHdG and F4/80 staining following DMM. CONCLUSION NOX4 deficiency restores cartilage homoeostasis, inhibits oxidative stress, inflammation and delays OA progression after DMM in mice. These findings suggest that NOX4 represent a potential target to counteract for OA treatment.
Collapse
Affiliation(s)
- Félix Renaudin
- Department of Cell Physiology and Metabolism, Université de Genève Faculté de médecine, Geneve, Switzerland.,Department of Orthopaedic Surgery, Geneva University Hospitals, Geneve, Switzerland
| | - Karim Oudina
- Department of Cell Physiology and Metabolism, Université de Genève Faculté de médecine, Geneve, Switzerland.,Department of Orthopaedic Surgery, Geneva University Hospitals, Geneve, Switzerland
| | - Maude Gerbaix
- Service of Bone Diseases, Department of Medicine, Geneva University Hospitals, Geneve, Switzerland
| | - Manon McGilligan Subilia
- Department of Cell Physiology and Metabolism, Université de Genève Faculté de médecine, Geneve, Switzerland.,Department of Orthopaedic Surgery, Geneva University Hospitals, Geneve, Switzerland
| | - Joris Paccaud
- Department of Cell Physiology and Metabolism, Université de Genève Faculté de médecine, Geneve, Switzerland.,Department of Orthopaedic Surgery, Geneva University Hospitals, Geneve, Switzerland
| | - Vincent Jaquet
- Department of Pathology and Immunology, Université de Genève Faculté de médecine, Geneve, Switzerland
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, Université de Genève Faculté de médecine, Geneve, Switzerland
| | - Serge Ferrari
- Service of Bone Diseases, Department of Medicine, Geneva University Hospitals, Geneve, Switzerland
| | - Thomas Laumonier
- Department of Cell Physiology and Metabolism, Université de Genève Faculté de médecine, Geneve, Switzerland.,Department of Orthopaedic Surgery, Geneva University Hospitals, Geneve, Switzerland
| | - Didier Hannouche
- Department of Cell Physiology and Metabolism, Université de Genève Faculté de médecine, Geneve, Switzerland .,Department of Orthopaedic Surgery, Geneva University Hospitals, Geneve, Switzerland
| |
Collapse
|
92
|
Zhang Q, Sun C, Liu X, Zhu C, Ma C, Feng R. Mechanism of immune infiltration in synovial tissue of osteoarthritis: a gene expression-based study. J Orthop Surg Res 2023; 18:58. [PMID: 36681837 PMCID: PMC9862811 DOI: 10.1186/s13018-023-03541-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/13/2023] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Osteoarthritis is a chronic degenerative joint disease, and increasing evidences suggest that the pathogenic mechanism involves immune system and inflammation. AIMS The aim of current study was to uncover hub genes linked to immune infiltration in osteoarthritis synovial tissue using comprehensive bioinformatics analysis and experimental confirmation. METHODS Multiple microarray datasets (GSE55457, GSE55235, GSE12021 and GSE1919) for osteoarthritis in Gene Expression Omnibus database were downloaded for analysis. Differentially expressed genes (DEGs) were identified using Limma package in R software, and immune infiltration was evaluated by CIBERSORT algorithm. Then weighted gene co-expression network analysis (WGCNA) was performed to uncover immune infiltration-associated gene modules. Protein-protein interaction (PPI) network was constructed to select the hub genes, and the tissue distribution of these genes was analyzed using BioGPS database. Finally, the expression pattern of these genes was confirmed by RT-qPCR using clinical samples. RESULTS Totally 181 DEGs between osteoarthritis and normal control were screened. Macrophages, mast cells, memory CD4 T cells and B cells accounted for the majority of immune cell composition in synovial tissue. Osteoarthritis synovial showed high abundance of infiltrating resting mast cells, B cells memory and plasma cells. WGCNA screened 93 DEGs related to osteoarthritis immune infiltration. These genes were involved in TNF signaling pathway, IL-17 signaling pathway, response to steroid hormone, glucocorticoid and corticosteroid. Ten hub genes including MYC, JUN, DUSP1, NFKBIA, VEGFA, ATF3, IL-6, PTGS2, IL1B and SOCS3 were selected by using PPI network. Among them, four genes (MYC, JUN, DUSP1 and NFKBIA) specifically expressed in immune system were identified and clinical samples revealed consistent change of these four genes in synovial tissue retrieved from patients with osteoarthritis. CONCLUSION A 4-gene-based diagnostic model was developed, which had well predictive performance in osteoarthritis. MYC, JUN, DUSP1 and NFKBIA might be biomarkers and potential therapeutic targets in osteoarthritis.
Collapse
Affiliation(s)
- Qingyu Zhang
- grid.460018.b0000 0004 1769 9639Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021 Shandong China
| | - Chao Sun
- grid.460018.b0000 0004 1769 9639Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021 Shandong China
| | - Xuchang Liu
- grid.460018.b0000 0004 1769 9639Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021 Shandong China
| | - Chao Zhu
- grid.460018.b0000 0004 1769 9639Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021 Shandong China
| | - Chuncheng Ma
- grid.460018.b0000 0004 1769 9639Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021 Shandong China
| | - Rongjie Feng
- grid.460018.b0000 0004 1769 9639Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Road Jing Wu Wei Qi, Jinan, 250021 Shandong China
| |
Collapse
|
93
|
Osthole Inhibits M1 Macrophage Polarization and Attenuates Osteolysis in a Mouse Skull Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:2975193. [PMID: 36686380 PMCID: PMC9851800 DOI: 10.1155/2023/2975193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 01/15/2023]
Abstract
Excessive bone resorption due to increased inflammatory factors is a common feature of inflammatory lytic bone diseases. This group of diseases is effectively treated with drugs. In recent years, many studies have reported that traditional Chinese medicine herbs have substantial effects on inflammation, osteoclast differentiation and maturation, and bone destruction. Herein, we investigated the effects of osthole (OST) on lipopolysaccharide- (LPS-) induced macrophage polarization, inflammatory responses, and osteolysis. In vitro, we used immunofluorescence and quantitative real-time polymerase chain reaction assays to confirm whether bone marrow-derived macrophages showed an increased expression of inflammatory factors, such as interleukin-6, iNOS, CCR7, and CD86, in the presence of LPS. However, we found that such expression was suppressed and that the M2 macrophage expression increased in the presence of OST. OST reduced LPS- and RANKL-induced intracellular reactive oxygen species production in the bone marrow-derived macrophages. Further, it potently suppressed osteoclast differentiation and osteoclast-specific gene expression by suppressing the P38/MAPK and NF-κB pathways. Consistent with the in vitro observations, OST greatly ameliorated LPS-induced bone resorption and modulated the ratio of macrophages at the site of osteolysis. Taken together, OST has great potential for use in the management of osteolytic diseases.
Collapse
|
94
|
He Y, Peng L, Xiong H, Liu W, Zhang H, Peng X, Zhu X, Guo F, Sun Y. The profiles of durian (Durio zibethinus Murr.) shell phenolics and their antioxidant effects on H 2O 2-treated HepG2 cells as well as the metabolites and organ distribution in rats. Food Res Int 2023; 163:112122. [PMID: 36596090 DOI: 10.1016/j.foodres.2022.112122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/28/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Durian is a nutritious tropical fruit with potent antioxidant, anti-inflammatory, antibacterial and anti-cancer effects. However, the durian shell was mainly discarded as waste, while there were few studies on the characterization of its phenolic profiles, antioxidant activities, and in vivo metabolites. In the present study, a total of 17 compounds were identified in durian shell extract (DSE) by using an ultra-high-performance liquid chromatography coupled with linear ion trap quadrupole Orbitrap mass spectrometry (UHPLC-LTQ-Orbitrap-MS/MS), while 33 metabolites were found in rats' plasma, urine and organ. Moreover, DSE could effectively reduce H2O2-induced oxidative damage in HepG2 cells, reduce the expression of Reactive Oxygen Species (ROS), Malondialdehyde (MDA) and Lactate Dehydrogenase (LDH) and inhibit apoptosis by regulating the expression of Bcl-2-Associated X (BAX), B-Cell Lymphoma 2 (BCL-2), Caspase-3 and Caspase-9 genes and proteins related to mitochondrial pathway apoptosis. This is the first comprehensive report on Durian shell phenolics, their metabolic profiles and underlying mechanisms of the in vitro antioxidant activities.
Collapse
Affiliation(s)
- Yangzheng He
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Li Peng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Hua Xiong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Wenqiang Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Hua Zhang
- Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, Jiangxi, PR China
| | - Xin Peng
- Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, Ningbo 315010, PR China
| | - Xuemei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Fanghua Guo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, PR China
| | - Yong Sun
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, PR China.
| |
Collapse
|
95
|
A Network Pharmacology-Based Study of Potential Targets of Angelicae Pubescentis-Herba Taxilli Compound for the Treatment of Osteoarthritis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4286168. [PMID: 36619785 PMCID: PMC9814887 DOI: 10.1155/2022/4286168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/09/2022] [Indexed: 12/30/2022]
Abstract
Objective Using network pharmacology and molecular docking, we explored the mechanism of Angelicae Pubescentis- (AP-) Herba Taxilli (HT) in the treatment of osteoarthritis (OA). Methods We selected Traditional Chinese Medicine Systems Pharmacology platform (TCMSP) to filtrate the practical components and targets of AP-HT. The disease targets of "osteoarthritis (OA)" were collected by GeneCards, DrugBank, TTD, OMIM, and PharmGKB databases, and the component-target interaction network was established by Cytoscape 3.9.1. Then, we set the protein-protein interaction (PPI) network by the STRING platform and visualized by Cytoscape 3.9.1. We also conducted Gene Ontology (GO) analysis and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis via the bioinformatics platform. Finally, we performed molecular docking using PyMOL 2.3.0 and AutoDock Vina software. Results 11 potential compounds were selected, and 1007 OA disease targets were collected. Ninety-four main targets of the AP-HT compound in the treatment of OA have been defined. PPI network demonstrated that JUN, RELA, TNF, IL6, MAPK1, TP53, AKT1, FOS, IL10, and MYC might serve as the critical targets of AP-HT for the treatment of OA. Moreover, membrane raft, membrane microdomain, cellular response to chemical stress, and cytokine receptor binding may play essential roles in the treatment of OA via GO analysis. The main functional pathways involved in these critical targets include fluid shear stress and atherosclerosis, lipid and atherosclerosis, IL-17 signaling pathway, age-range signaling pathway in diabetic composites, and TNF signaling pathway via KEGG analysis. The results of molecular docking showed that the critical ingredients of AP-HT had an excellent affinity to related nuclear genes.
Collapse
|
96
|
Gambari L, Cellamare A, Grassi F, Grigolo B, Panciera A, Ruffilli A, Faldini C, Desando G. Overview of Anti-Inflammatory and Anti-Nociceptive Effects of Polyphenols to Halt Osteoarthritis: From Preclinical Studies to New Clinical Insights. Int J Mol Sci 2022; 23:ijms232415861. [PMID: 36555503 PMCID: PMC9779856 DOI: 10.3390/ijms232415861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Knee osteoarthritis (OA) is one of the most multifactorial joint disorders in adults. It is characterized by degenerative and inflammatory processes that are responsible for joint destruction, pain and stiffness. Despite therapeutic advances, the search for alternative strategies to target inflammation and pain is still very challenging. In this regard, there is a growing body of evidence for the role of several bioactive dietary molecules (BDMs) in targeting inflammation and pain, with promising clinical results. BDMs may be valuable non-pharmaceutical solutions to treat and prevent the evolution of early OA to more severe phenotypes, overcoming the side effects of anti-inflammatory drugs. Among BDMs, polyphenols (PPs) are widely studied due to their abundance in several plants, together with their benefits in halting inflammation and pain. Despite their biological relevance, there are still many questionable aspects (biosafety, bioavailability, etc.) that hinder their clinical application. This review highlights the mechanisms of action and biological targets modulated by PPs, summarizes the data on their anti-inflammatory and anti-nociceptive effects in different preclinical in vitro and in vivo models of OA and underlines the gaps in the knowledge. Furthermore, this work reports the preliminary promising results of clinical studies on OA patients treated with PPs and discusses new perspectives to accelerate the translation of PPs treatment into the clinics.
Collapse
Affiliation(s)
- Laura Gambari
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Antonella Cellamare
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Francesco Grassi
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Alessandro Panciera
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Alberto Ruffilli
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Cesare Faldini
- 1st Orthopedic and Traumatology Clinic, IRCCS Istituto Ortopedico Rizzoli, via G.C. Pupilli 1, 40136 Bologna, Italy
| | - Giovanna Desando
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, via di Barbiano 1/10, 40136 Bologna, Italy
- Correspondence: ; Tel.: +39-0516366803
| |
Collapse
|
97
|
Sartinah A, Nugrahani I, Ibrahim S, Anggadiredja K. Potential metabolites of Arecaceae family for the natural anti-osteoarthritis medicine: A review. Heliyon 2022; 8:e12039. [PMID: 36561673 PMCID: PMC9763769 DOI: 10.1016/j.heliyon.2022.e12039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/28/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a chronic inflammatory disorder of the joints caused by fluid and cartilage matrix component reduction. This disease results in symptoms of pain, deformity, and limitation of movement. In general, OA is treated with anti-inflammatory drugs and chondroprotection compounds, includes natural nutraceutical ingredients, which are expected to be effective and have minimal side effects. Arecaceae plants are widely spread worldwide, especially in tropical areas. The objective of this review is to collect information about the Arecaceae family as anti-OA agents, with the main study focusing on the primary and secondary metabolites of plants of the Arecaceae family, i.e., sugar palm (Arenga pinnata), nipa palm (Nypa fruticans), palmyra palm (Borassus flabellifer), date palm (Phoenix dactylifera), and betel nut (Areca catechu) have potential as anti-OA agents. The Arecaceae's metabolites that show anti-inflammatory and chondroprotective effects are galactomannan, fatty acids (linoleic and linolenic acids), flavonoids (quercetin, luteolin, isorhamnetin), phenolics (coumaric acid, ferulic acid), polyphenols (epicatechin), and steroids (stigmasterol, campesterol, spirostane). Based on the reports, the Arecaceae family plants become worthy of being explored and developed into natural anti-OA products, such as supplements or nutraceuticals.
Collapse
Affiliation(s)
- Ari Sartinah
- School of Pharmacy, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Ilma Nugrahani
- School of Pharmacy, Bandung Institute of Technology, Bandung 40132, Indonesia
- Corresponding author.
| | - Slamet Ibrahim
- Faculty of Pharmacy, Universitas Jenderal Achmad Yani, Cimahi, Indonesia
| | | |
Collapse
|
98
|
Liu Y, Zhang Z, Li T, Xu H, Zhang H. Senescence in osteoarthritis: from mechanism to potential treatment. Arthritis Res Ther 2022; 24:174. [PMID: 35869508 PMCID: PMC9306208 DOI: 10.1186/s13075-022-02859-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is an age-related cartilage degenerative disease, and chondrocyte senescence has been extensively studied in recent years. Increased numbers of senescent chondrocytes are found in OA cartilage. Selective clearance of senescent chondrocytes in a post-traumatic osteoarthritis (PTOA) mouse model ameliorated OA development, while intraarticular injection of senescent cells induced mouse OA. However, the means and extent to which senescence affects OA remain unclear. Here, we review the latent mechanism of senescence in OA and propose potential therapeutic methods to target OA-related senescence, with an emphasis on immunotherapies. Natural killer (NK) cells participate in the elimination of senescent cells in multiple organs. A relatively comprehensive discussion is presented in that section. Risk factors for OA are ageing, obesity, metabolic disorders and mechanical overload. Determining the relationship between known risk factors and senescence will help elucidate OA pathogenesis and identify optimal treatments.
Collapse
|
99
|
Anti-Inflammatory and Chondroprotective Effects Induced by Phenolic Compounds from Onion Waste Extracts in ATDC-5 Chondrogenic Cell Line. Antioxidants (Basel) 2022; 11:antiox11122381. [PMID: 36552589 PMCID: PMC9774380 DOI: 10.3390/antiox11122381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Osteoarthritis is a prevalent degenerative condition that is closely related to the destruction and inflammation of cartilage. The high prevalence of this pathology exhorts researchers to search for novel therapeutic approaches. Vegetable-fruit wastes have emerged as a promising origin of anti-inflammatory and antioxidant compounds that, in some cases, may also exert chondroprotective effects. This study aims to decipher the potential of onion waste products in the inhibition of molecular events involved in osteoarthritis. Onion extracts showed a high content of phenolic compounds and antioxidant properties. Cytocompatibility was demonstrated in the chondrogenic cell line ATDC-5, exerting viability percentages higher than 90% and a slight increase in the S phase cycle cell. The induction of inflammation mediated by the lipopolysaccharide and onion extracts' treatment substantially inhibited molecular markers related to inflammation and cartilage degradation, highlighting the promising application of onion extracts in biomedical approaches. The in silico analyses suggested that the results could be attributed to protocatechuic, ellagic, and vanillic acids' greater cell membrane permeability. Our work provides distinctive information about the possible application of waste onion extracts as functional components with anti-inflammatory and chondroprotective characteristics in osteoarthritis.
Collapse
|
100
|
Xu B, Huang W. Effect and mechanisms of quercetin on the treatment of osteoarthritis: A preliminary pre-clinical study. Asian J Surg 2022; 46:2132-2134. [PMID: 36428143 DOI: 10.1016/j.asjsur.2022.11.056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/11/2022] [Indexed: 11/24/2022] Open
|