1
|
Ye H, Li ZQ, Yang JM, Long Y, Zhong YB, Wu Y, Wang MY. A network pharmacology-based study to investigate the mechanism of curcumin-regulated regenerative repair of quadriceps femoris muscle in KOA rats. Eur J Pharmacol 2024; 982:176910. [PMID: 39154821 DOI: 10.1016/j.ejphar.2024.176910] [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: 05/05/2024] [Revised: 07/29/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a very common musculoskeletal disorder, and patients with KOA often exhibit significant quadriceps femoris muscle atrophy. It is well established that curcumin (CUR) exerts protective effects on skeletal muscle. However, the efficacy of CUR in treating KOA-induced quadriceps femoris muscle atrophy and its underlying mechanisms remain uncertain. In this study, we employed network pharmacology to investigate the mechanism by which CUR promotes regenerative repair of the quadriceps femoris muscle in rats with KOA. METHODS The potential targets of CUR were obtained from Swiss Target Prediction. The targets of skeletal muscle regeneration were identified from GeneCard and OMIM. A Venn diagram was generated to visualize the intersection of CUR targets and skeletal muscle regeneration targets, and the core targets were identified using STRING. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses were conducted using DAVID. Finally, the network pharmacology results were further validated by establishing a KOA rat model using the Hulth method. RESULTS Network pharmacology analysis and molecular docking results revealed that CUR affects skeletal muscle regeneration through multiple targets and pathways. In vivo experimental results were validated by demonstrating that KOA causes atrophy and induces apoptosis in the quadriceps femoris muscle. Furthermore, CUR was shown to inhibit apoptosis in the quadriceps femoris muscle by regulating STAT3 and FOS, as well as the PI3K/AKT signaling pathway. CONCLUSIONS Our study revealed the apoptosis-inhibiting effects of CUR and its underlying mechanisms. Consequently, CUR has the potential to improve quadriceps femoris muscle atrophy caused by KOA.
Collapse
Affiliation(s)
- Hua Ye
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Ze-Qin Li
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Jia-Ming Yang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Yi Long
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Gannan Medical University, Ganzhou City, Jiangxi, China
| | - Yan-Biao Zhong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou City, Jiangxi, China
| | - Yi Wu
- Gannan Medical University, Ganzhou City, Jiangxi, China; Jiangxi Provincal Key Laboratory of Tissue Engineering (2024SSY06291), School of Pharmacy, Gannan Medical University, Gouzhou, Jiangxi, China.
| | - Mao-Yuan Wang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi, China; Ganzhou Key Laboratory of Rehabilitation Medicine, GanZhou City, Jiangxi, China.
| |
Collapse
|
2
|
Liu K, Zhang B, Zhang X. Promoting Articular Cartilage Regeneration through Microenvironmental Regulation. J Immunol Res 2024; 2024:4751168. [PMID: 39104594 PMCID: PMC11300091 DOI: 10.1155/2024/4751168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
In recent years, as the aging population continues to grow, osteoarthritis (OA) has emerged as a leading cause of disability, with its incidence rising annually. Current treatments of OA include exercise and medications in the early stages and total joint replacement in the late stages. These approaches only relieve pain and reduce inflammation; however, they have significant side effects and high costs. Therefore, there is an urgent need to identify effective treatment methods that can delay the pathological progression of this condition. The changes in the articular cartilage microenvironment, which are complex and diverse, can aggravate the pathological progression into a vicious cycle, inhibiting the repair and regeneration of articular cartilage. Understanding these intricate changes in the microenvironment is crucial for devising effective treatment modalities. By searching relevant research articles and clinical trials in PubMed according to the keywords of articular cartilage, microenvironment, OA, mechanical force, hypoxia, cytokine, and cell senescence. This study first summarizes the factors affecting articular cartilage regeneration, then proposes corresponding treatment strategies, and finally points out the future research direction. We find that regulating the opening of mechanosensitive ion channels, regulating the expression of HIF-1, delivering growth factors, and clearing senescent cells can promote the formation of articular cartilage regeneration microenvironment. This study provides a new idea for the treatment of OA in the future, which can promote the regeneration of articular cartilage through the regulation of the microenvironment so as to achieve the purpose of treating OA.
Collapse
Affiliation(s)
- Kai Liu
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and MinistryGuangxi Medical University, Nanning, Guangxi 530021, China
| | - Bingjun Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaoling Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and MinistryGuangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
3
|
Chen T, Jin L, Li J, Liu Y. Pyroptosis mediates osteoporosis via the inflammation immune microenvironment. Front Immunol 2024; 15:1371463. [PMID: 38895114 PMCID: PMC11184911 DOI: 10.3389/fimmu.2024.1371463] [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: 01/16/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoporosis represents a systemic imbalance in bone metabolism, augmenting the susceptibility to fractures among patients and emerging as a notable mortality determinant in the elderly population. It has evolved into a worldwide concern impacting the physical well-being of the elderly, imposing a substantial burden on both human society and the economy. Presently, the precise pathogenesis of osteoporosis remains inadequately characterized and necessitates further exploration. The advancement of osteoporosis is typically linked to the initiation of an inflammatory response. Cells in an inflammatory environment can cause inflammatory death including pyroptosis. Pyroptosis is a recently identified form of programmed cell death with inflammatory properties, mediated by the caspase and gasdermin families. It is regarded as the most inflammatory form of cell death in contemporary medical research. Under the influence of diverse cytokines, macrophages, and other immune cells may undergo pyroptosis, releasing inflammatory factors, such as IL-1β and IL-18. Numerous lines of evidence highlight the pivotal role of pyroptosis in the pathogenesis of inflammatory diseases, including cancer, intestinal disorders, hepatic conditions, and cutaneous ailments. Osteoporosis progression is frequently associated with inflammation; hence, pyroptosis may also play a role in the pathogenesis of osteoporosis to a certain extent, making it a potential target for treatment. This paper has provided a comprehensive summary of pertinent research concerning pyroptosis and its impact on osteoporosis. The notion proposing that pyroptosis mediates osteoporosis via the inflammatory immune microenvironment is advanced, and we subsequently investigate potential targets for treating osteoporosis through the modulation of pyroptosis.
Collapse
Affiliation(s)
- Te Chen
- Division of Joint Surgery, Department of Orthopaedics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Linyu Jin
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingyi Li
- Division of Joint Surgery, Department of Orthopaedics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yikai Liu
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
4
|
Ye H, Long Y, Yang JM, Wu YL, Dong LY, Zhong YB, Luo Y, Wang MY. Curcumin regulates autophagy through SIRT3-SOD2-ROS signaling pathway to improve quadriceps femoris muscle atrophy in KOA rat model. Sci Rep 2024; 14:8176. [PMID: 38589505 PMCID: PMC11001965 DOI: 10.1038/s41598-024-58375-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/28/2024] [Indexed: 04/10/2024] Open
Abstract
Knee osteoarthritis (KOA) usually leads to quadriceps femoris atrophy, which in turn can further aggravate the progression of KOA. Curcumin (CUR) has anti-inflammatory and antioxidant effects and has been shown to be a protective agent for skeletal muscle. CUR has been shown to have a protective effect on skeletal muscle. However, there are no studies related to whether CUR improves KOA-induced quadriceps femoris muscle atrophy. We established a model of KOA in rats. Rats in the experimental group were fed CUR for 5 weeks. Changes in autophagy levels, reactive oxygen species (ROS) levels, and changes in the expression of the Sirutin3 (SIRT3)-superoxide dismutase 2 (SOD2) pathway were detected in the quadriceps femoris muscle of rats. KOA led to quadriceps femoris muscle atrophy, in which autophagy was induced and ROS levels were increased. CUR increased SIRT3 expression, decreased SOD2 acetylation and ROS levels, inhibited the over-activation of autophagy, thereby alleviating quadriceps femoris muscle atrophy and improving KOA. CUR has a protective effect against quadriceps femoris muscle atrophy, and KOA is alleviated after improvement of quadriceps femoris muscle atrophy, with the possible mechanism being the reduction of ROS-induced autophagy via the SIRT3-SOD2 pathway.
Collapse
Affiliation(s)
- Hua Ye
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Yi Long
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Jia-Ming Yang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Yan-Lin Wu
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Ling-Yan Dong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Yan-Biao Zhong
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China
- Ganzhou Intelligent Rehabilitation Technology Innovation Center, Ganzhou, Jiangxi, China
| | - Yun Luo
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
| | - Mao-Yuan Wang
- Department of Rehabilitation Medicine, First Affiliated Hospital of Gannan Medical University, 128 Jinling Road, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
- Ganzhou Key Laboratory of Rehabilitation Medicine, Ganzhou, Jiangxi, China.
| |
Collapse
|
5
|
Jiang D, Cheng S, Kang P, Li T, Li X, Xiao J, Ren L. microRNA-105-5p protects against chondrocyte injury, extracellular matrix degradation, and osteoarthritis progression by targeting SPARCL1. Histol Histopathol 2024; 39:483-496. [PMID: 37534667 DOI: 10.14670/hh-18-654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
OBJECTIVE Both microRNA (miR)-105-5p and SPARCL1 were discovered to be differentially expressed in osteoarthritis (OA), but their roles and exact mechanisms have not been entirely elaborated. This paper sets out to probe the impact of miR-105-5p/SPARCL1 on chondrocyte injury, extracellular matrix degradation, and osteoarthritis progression. METHODS C28/I2 cells were stimulated with IL-1β to construct an in vitro OA model. C28/I2 cells were transfected with sh-SPARCL1, oe-SPARCL1, or miR-105-5p mimic before IL-1β induction. CCK-8 assay, flow cytometry, and ELISA were adopted to assess cell viability, apoptosis, and inflammatory factor expression, respectively. The binding relationship of miR-105-5p to SPARCL1 was assessed using dual-luciferase reporter assay. After an OA rat model was established, rats underwent intra-articular injection with ago-miR-105-5p. TUNEL was applied to determine cell apoptosis in vivo. mRNA and protein levels were measured by qRT-PCR and western blot, respectively, in vitro and in vivo. RESULTS IL-1β treatment diminished miR-105-5p expression and augmented SPARCL1 expression in C28/I2 cells. miR-105-5p decreased SPARCL1 expression by targeting SPARCL1. miR-105-5p overexpression or SPARCL1 silencing prominently reversed the decrease in viability and the promotion of inflammatory factor production, cartilage matrix degradation, and apoptosis in IL-1β-stimulated C28/I2 cells. Furthermore, upregulation of SPARCL1 nullified the influence of miR-105-5p overexpression on viability, apoptosis, inflammation, and cartilage matrix degradation in IL-1β-stimulated C28/I2 cells. miR-105-5p overexpression ameliorated knee cartilage tissue injury in OA rats. CONCLUSION Conclusively, miR-105-5p exerted suppressive effects on chondrocyte injury, extracellular matrix degradation, and OA progression by targeting SPARCL1.
Collapse
Affiliation(s)
- Dong Jiang
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China
| | - Shigao Cheng
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China
| | - Pengcheng Kang
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China
| | - Tengfei Li
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China
| | - Xun Li
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China
| | - Jiongzhe Xiao
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China
| | - Lian Ren
- Department of Orthopedic Surgery, Loudi Central Hospital, Loudi, Hunan, PR China.
| |
Collapse
|
6
|
Truchan K, Osyczka AM. Noggin promotes osteogenesis in human adipose-derived mesenchymal stem cells via FGFR2/Src/Akt and ERK signaling pathway. Sci Rep 2024; 14:6724. [PMID: 38509118 PMCID: PMC10954655 DOI: 10.1038/s41598-024-56858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The balance between Noggin and bone morphogenetic proteins (BMPs) is important during early development and skeletal regenerative therapies. Noggin binds BMPs in the extracellular space, thereby preventing BMP signaling. However, Noggin may affect cell response not necessarily through the modulation of BMP signaling, raising the possibility of direct Noggin signaling through yet unspecified receptors. Here we show that in osteogenic cultures of adipose-derived stem cells (ASCs), Noggin activates fibroblast growth factor receptors (FGFRs), Src/Akt and ERK kinases, and it stabilizes TAZ proteins in the presence of dexamethasone. Overall, this leads ASCs to increased expression of osteogenic markers and robust mineral deposition. Our results also indicate that Noggin can induce osteogenic genes expression in normal human bone marrow stem cells and alkaline phosphatase activity in normal human dental pulp stem cells. Besides, Noggin can specifically activate FGFR2 in osteosarcoma cells. We believe our findings open new research avenues to further explore the involvement of Noggin in cell fate modulation by FGFR2/Src/Akt/ERK signaling and potential applications of Noggin in bone regenerative therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| |
Collapse
|
7
|
Han X, Bai F, Li P, Bai X, Zhang Y, Wang W. Identification of novel potential drugs for the treatment and prevention of osteoarthritis. Biochem Biophys Rep 2024; 37:101647. [PMID: 38304574 PMCID: PMC10830515 DOI: 10.1016/j.bbrep.2024.101647] [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: 10/23/2023] [Revised: 12/27/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024] Open
Abstract
Objectives Osteoarthritis (OA) is characterized by a high prevalence, poor prognosis, and a propensity to lead to disability. Despite the availability of standard therapies, they are associated with potential side effects and don't provide a complete cure for patients. Consequently, there is an urgent demand for the development of novel drugs. Method The gene expression profiles (GSE64394, GSE178557 and GSE215039) of normal and OA chondrocytes samples were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were identified by the "LIMMA" R package. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment were conducted using the R package clusterProfiler. A protein-protein (PPI) interaction network was performed to identify hub genes by using the Search Tool for the Retrieval of Interacting Genes (STRING) and Cytoscape. Small molecule compounds linked to OA were predicted through the NetworkAnalyst platform. Finally, molecular docking was conducted using AutoDock and Pymol software. Results We identified 98 DEGs primarily implicated in endochondral ossification, extracellular matrix degradation, and Wnt signaling pathways. 23 DEGs were closely associated with OA, and 10 hub genes were found to be potential drug targets for OA. Two new targeted compounds, tetrachlorodibenzodioxin (TCDD) and valproic acid (VPA), were screened. And they both exhibited strong binding affinity to their respective targets. Conclusions Reducing exposure to TCDD could be a crucial strategy in preventing OA, and VPA has gained recognition as a novel drug candidate for OA treatment.
Collapse
Affiliation(s)
- Xiaosong Han
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Fan Bai
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Peng Li
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Xiaojin Bai
- Department of Osteology, the First People's Hospital of Zunyi, Zunyi, Guizhou, 563099, China
| | - Yanli Zhang
- Biological Medicine Research and Development Center, Yangtze Delta of Zhejiang, Hangzhou, Zhejiang, 314006, China
| | - Wenmin Wang
- Biological Medicine Research and Development Center, Yangtze Delta of Zhejiang, Hangzhou, Zhejiang, 314006, China
| |
Collapse
|
8
|
Huang H, Lin X, Meng X, Liu Y, Fan J, Zhu L, Chen J, Zhang L, Mi H, Deng J. Effects of replacing wheat bran with palm kernel cake or fermented palm kernel cake on the growth performance, intestinal microbiota and intestinal health of tilapia (GIFT, Oreochromis niloticus). Front Nutr 2024; 11:1368251. [PMID: 38450228 PMCID: PMC10915778 DOI: 10.3389/fnut.2024.1368251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/12/2024] [Indexed: 03/08/2024] Open
Abstract
A nine-week feeding trial was conducted to evaluate the effects of replacing wheat bran (WB) with palm kernel cake (PKC) or fermented palm kernel cake (FPKC) on the growth performance, intestinal microbiota and intestinal health of genetically improved farmed tilapia (GIFT, Oreochromis niloticus) (initial weight 7.00 ± 0.01 g). Eleven isonitrogenous and isolipidic experimental diets were formulated by replacing 0, 20, 40, 60, 80, and 100% of dietary WB with PKC or FPKC. Replacement of WB with PKC concentrations up to 80% had no significant effect on the growth rate of tilapia or feed utilisation (p > 0.05). FPKC improved the growth performance of tilapia, with optimum growth achieved at 40% replacement level (p < 0.05). Complete replacement with PKC significantly decreased the activity of lipase and trypsin, and reduced the height of muscularis and the height of villus (p < 0.05). However, FPKC significantly increased amylase activity and villus height (p < 0.05). The apparent digestibility of dry matter and energy decreased linearly with increasing levels of PKC substitution, while FPKC showed the opposite trend (p < 0.05). PKC replacement of WB by 20% significantly reduced serum diamine oxidase activity and endothelin levels and increased intestinal tight junctions (p < 0.05). However, FPKC significantly decreased diamine oxidase activity and increased intestinal tight junctions (p < 0.05). PKC completely replaced WB, up-regulating the expression of pro-inflammatory factors (il-1β) (p < 0.05). When 40% of WB was replaced with FPKC, the expression of pro-inflammatory factors (il-1β and il-6) was decreased significantly (p < 0.05). Completely replacement of WB with PKC reduced the abundance of Firmicutes and Chloroflexi, while FPKC reduced the abundance of Fusobacteriota and increased the levels of Actinobacteriota. WB can be replaced with PKC up to 80% in tilapia feeds. However, the high percentage of gluten induced intestinal inflammation, impaired gut health, and reduced dietary nutrient utilisation and growth performance. Complete replacement of WB with FPKC promoted intestinal immunity. It also improved dietary nutrient utilisation and growth performance. However, the optimal growth was achieved at a 40% replacement level.
Collapse
Affiliation(s)
- Huajing Huang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Xiangqin Lin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Xiaoxue Meng
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yu Liu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiongting Fan
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Lulu Zhu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Jiaxian Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Lu Zhang
- Tongwei Agricultural Development Co., Ltd., Chengdu, Sichuan, China
| | - Haifeng Mi
- Tongwei Agricultural Development Co., Ltd., Chengdu, Sichuan, China
| | - Junming Deng
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
- Aquatic Animals Precision Nutrition and High-Efficiency Feed Engineering Research Centre of Guangdong Province, Guangdong Ocean University, Zhanjiang, Guangdong, China
| |
Collapse
|
9
|
Li H, Huang L, Zhao R, Wu G, Yin Y, Zhang C, Li P, Guo L, Wei X, Che X, Li L. TSP-1 increases autophagy level in cartilage by upregulating HSP27 which delays progression of osteoarthritis. Int Immunopharmacol 2024; 128:111475. [PMID: 38183909 DOI: 10.1016/j.intimp.2023.111475] [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: 09/17/2023] [Revised: 12/22/2023] [Accepted: 12/29/2023] [Indexed: 01/08/2024]
Abstract
This study aimed to determine whether Thrombospondin-1 (TSP-1) can be used as a biomarker to diagnose early osteoarthritis (OA) and whether it has a chondroprotective effect against OA. We examined TSP-1 expression in cartilage, synovial fluid, and serum at different time points after anterior cruciate ligament transection (ACLT) surgery in rats. Subsequently, TSP-1 was overexpressed or silenced to detect its effects on extracellular matrix (ECM) homeostasis, autophagy level, proliferation and apoptosis in chondrocytes. Adenovirus encoding TSP-1 was injected into the knee joints of ACLT rats to test its effect against OA. Combined with proteomic analysis, the molecular mechanism of TSP-1 in cartilage degeneration was explored. Intra-articular injection of an adenovirus carrying the TSP-1 sequence showed chondroprotective effects against OA. Moreover, TSP-1 expression decreases with OA progression and can effectively promote cartilage proliferation, inhibit apoptosis, and helps to sustain the balance between ECM anabolism and catabolism. Overexpression of TSP-1 also can increase autophagy by upregulating Heat Shock Protein 27 (HSP27, hspb1), thereby enhancing its effect as a stimulator of autophagy. TSP-1 is a hopeful strategy for OA treatment.
Collapse
Affiliation(s)
- Haoqian Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Lingan Huang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China; Department of Sports Medicine Center, Beijing Tsinghua Changgung Hospital, Beijing, China
| | - Ruipeng Zhao
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Gaige Wu
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Yukun Yin
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Chengming Zhang
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Pengcui Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Li Guo
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Xianda Che
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Lu Li
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics , The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China.
| |
Collapse
|
10
|
Wang D, Zhang Z, Li X, He L. RNA binding protein PUM2 promotes IL-1β-induced apoptosis of chondrocytes via regulating FOXO3 expression. Heliyon 2024; 10:e25080. [PMID: 38356524 PMCID: PMC10865267 DOI: 10.1016/j.heliyon.2024.e25080] [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: 07/13/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Objective RNA-binding proteins (RBPs) have been recently proven to be involved in the pathogenesis of several diseases. However, few studies elaborated RBPs in regulating osteoarthritis. This study aims to define the function and mechanism of RBPs-PUM2 in chondrocyte apoptosis during osteoarthritis. Methods Cartilage tissue samples and human juvenile chondrocyte cell line C28/I2 were collected for further study. PUM2 expression in the human tissues and cells was determined using qRT-PCR. Chondrocyte viability and apoptosis were determined by MTT and flow cytometry. ROS generation was determined by flow cytometry. The regulation of PUM2 on FOXO3 translation was evaluated by RNA immunoprecipitation, RNA pull-down, and Luciferase gene reporter analysis. Results PUM2 is upregulated in both cartilage tissue of osteoarthritis patients and IL-1β-stimulated chondrocytes. PUM2 overexpression reduces cell viability and promotes cell apoptosis and ROS generation of chondrocytes. PUM2 silencing increases cell viability and ameliorates cell apoptosis as well as ROS generation in chondrocytes induced by IL-1β. PUM2 inhibits FOXO3 expression via binding its mRNA 3'-UTR. PUM2 forms a signaling axis with FOXO3 in IL-1β induced chondrocyte damage. Conclusion PUM2 is upregulated in cartilage tissue of osteoarthritis and positively regulates chondrocytes apoptosis through controlling FOXO3 protein expression.
Collapse
Affiliation(s)
- Du Wang
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - ZhiLi Zhang
- Department of Surgery, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Xili Li
- Department of Radiology, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| | - Ling He
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Fan SH, Chang Y, Xiong XY, Xiang M, Yuan WL, Yang XQ, Wei WH, Chen L, Cheng MN, Zhu FH, He SJ, Zuo JP, Lin ZM. Reversible SAHH inhibitor ameliorates MIA-induced osteoarthritis of rats through suppressing MEK/ERK pathway. Biomed Pharmacother 2024; 170:115975. [PMID: 38070246 DOI: 10.1016/j.biopha.2023.115975] [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: 09/10/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 01/10/2024] Open
Abstract
Osteoarthritis (OA) is characterized by gradual articular cartilage degradation, accompanied by persistent low-grade joint inflammation, correlating with radiographic and pain-related progression. The latent therapeutic potential of DZ2002, a reversible inhibitor of S-adenosyl-L-homocysteine hydrolase (SAHH), holds promise for OA intervention. This study endeavored to examine the therapeutic efficacy of DZ2002 within the milieu of OA. The cytotoxicity of DZ2002 was evaluated using the MTT assay on bone marrow-derived macrophages. The inhibitory impact of DZ2002 during the process of osteoclastogenesis was assessed using TRAP staining, analysis of bone resorption pits, and F-actin ring formation. Mechanistic insights were derived from qPCR and Western blot analyses. Through the intra-articular injection of monosodium iodoacetate (MIA), an experimental rat model of OA was successfully instituted. This was subsequently accompanied by a series of assessments including Von Frey filament testing, analysis of weight-bearing behaviors, and micro-CT imaging, all aimed at assessing the effectiveness of DZ2002. The findings emphasized the effectiveness of DZ2002 in mitigating osteoclastogenesis induced by M-CSF/RANKL, evident through a reduction in TRAP-positive OCs and bone resorption. Moreover, DZ2002 modulated bone resorption-associated gene and protein expression (CTSK, CTR, Integrin β3) via the MEK/ERK pathway. Encouragingly, DZ2002 also alleviates MIA-induced pain, cartilage degradation, and bone loss. In conclusion, DZ2002 emerges as a potential therapeutic contender for OA, as evidenced by its capacity to hinder in vitro M-CSF/RANKL-induced osteoclastogenesis and mitigate in vivo osteoarthritis progression. This newfound perspective provides substantial support for considering DZ2002 as a compelling agent for osteoarthritis intervention.
Collapse
Affiliation(s)
- Shu-Hui Fan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuan Chang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao-Yu Xiong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mai Xiang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Long Yuan
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Qian Yang
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wen-Hui Wei
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Chen
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Meng-Nan Cheng
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Feng-Hua Zhu
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Shi-Jun He
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jian-Ping Zuo
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu Province 210029, China; Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China; Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Ze-Min Lin
- Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
12
|
Lyons KM, Rosen V. Strengthening the case for BMP inhibition as a point of intervention for osteoarthritis. Osteoarthritis Cartilage 2023; 31:1423-1424. [PMID: 37625479 DOI: 10.1016/j.joca.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023]
Affiliation(s)
- Karen M Lyons
- Department of Orthopaedic Surgery, University of California, Los Angeles, United States.
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, United States.
| |
Collapse
|
13
|
Jaswal AP, Kumar B, Roelofs AJ, Iqbal SF, Singh AK, Riemen AHK, Wang H, Ashraf S, Nanasaheb SV, Agnihotri N, De Bari C, Bandyopadhyay A. BMP signaling: A significant player and therapeutic target for osteoarthritis. Osteoarthritis Cartilage 2023; 31:1454-1468. [PMID: 37392862 DOI: 10.1016/j.joca.2023.05.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/25/2023] [Accepted: 05/18/2023] [Indexed: 07/03/2023]
Abstract
OBJECTIVE To explore the significance of BMP signaling in osteoarthritis (OA) etiology, and thereafter propose a disease-modifying therapy for OA. METHODS To examine the role of the BMP signaling in pathogenesis of OA, an Anterior Cruciate Ligament Transection (ACLT) surgery was performed to incite OA in C57BL/6J mouse line at postnatal day 120 (P120). Thereafter, to investigate whether activation of BMP signaling is necessary and sufficient to induce OA, we have used conditional gain- and loss-of-function mouse lines in which BMP signaling can be activated or depleted, respectively, upon intraperitoneal injection of tamoxifen. Finally, we locally inhibited BMP signaling through intra-articular injection of LDN-193189 pre- and post-onset surgically induced OA. The majority of the investigation has been conducted using micro-CT, histological staining, and immuno histochemistry to assess the disease etiology. RESULTS Upon induction of OA, depletion of SMURF1-an intra-cellular BMP signaling inhibitor in articular cartilage coincided with the activation of BMP signaling, as measured by pSMAD1/5/9 expression. In mouse articular cartilage, the BMP gain-of-function mutation is sufficient to induce OA even without surgery. Further, genetic, or pharmacological BMP signaling suppression also prevented pathogenesis of OA. Interestingly, inflammatory indicators were also significantly reduced upon LDN-193189 intra-articular injection which inhibited BMP signaling and slowed OA progression post onset. CONCLUSION Our findings showed that BMP signaling is crucial to the etiology of OA and inhibiting BMP signaling locally can be a potent strategy for alleviating OA.
Collapse
Affiliation(s)
- Akrit Pran Jaswal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Bhupendra Kumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Anke J Roelofs
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Sayeda Fauzia Iqbal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Amaresh Kumar Singh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
| | - Anna H K Riemen
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Hui Wang
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Sadaf Ashraf
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Sanap Vaibhav Nanasaheb
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Nitin Agnihotri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
14
|
Du XF, Huang K, Chen XY, Huang CH, Cao HY, Wang GJ, Hu Y. Gremlin-1 promotes IL-1β-stimulated chondrocyte inflammation and extracellular matrix degradation via activation of the MAPK signaling pathway. J Biochem Mol Toxicol 2023; 37:e23404. [PMID: 37352019 DOI: 10.1002/jbt.23404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/18/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023]
Abstract
The role and mechanism of Gremlin-1 in osteoarthritis (OA) were expected to be probed in this study. Firstly, an in vitro OA model was constructed by stimulating human chondrocyte cell line CHON-001 with IL-1β. Next, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) were utilized for assessing the effect of IL-1β with different concentrations (5, 10, and 20 ng/mL) on the activity and Gremlin-1 messenger RNA of CHON-001 cells, respectively. Besides, the influence of knocking down/over-expressing Gremlin-1 on the inflammatory factors (IL-6, TNF-α, IL-18 and PGE2), oxidative stress-related substances (malondialdehyde [MDA]; superoxide dismutase [SOD]; lactate dehydrogenase [LDH]), extracellular matrix (ECM) degradation-related proteins, and mitogen-activated protein kinase (MAPK) pathway proteins in IL-1β-stimulated CHON-001 cells were tested by enzyme-linked immunosorbent assay, related kits, qRT-PCR, and western blot, respectively. IL-1β inhibited CHON-001 cell proliferation and upregulated Gremlin-1 expression in a concentration-dependent manner. Overexpression of Gremlin-1 increased the IL-6, TNF-α, IL-18, PGE2, and MDA levels, enhanced the LDH activity, and decreased the SOD activity in IL-1β-induced CHON-001 cells; while the effect of Gremlin-1 knockdown on the above factors was in contrast with that of the overexpression. Furthermore, overexpression of Gremlin-1 upregulated protein expression of matrix metalloproteinase (MMP)-3, MMP-13, and ADAMTS4 while downregulated protein expression of collagen III, aggrecan, and SOX-9 in IL-1β-stimulated CHON-001 cells. Besides, overexpression of Gremlin-1 increased the p-p38/p38 value while decreased the p-JNK/JNK value in L-1β-stimulated CHON-001 cells; however, knockdown of Gremlin-1 reversed the above results. Gremlin-1 may promote IL-1β-stimulated CHON-001 cell inflammation and ECM degradation by activating the MAPK signaling pathway.
Collapse
Affiliation(s)
- Xiu-Fan Du
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Kai Huang
- Department of Orthopedics, Zibo Orthopaedic Hospital Shandong Province, Zibo, Shandong, China
| | - Xiao-Yan Chen
- Department of stomatology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Chun-Hang Huang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Hui-Yuan Cao
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Guang-Ji Wang
- Department of Sports Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yong Hu
- Department of Orthopedics, Danzhou People's Hospital, Danzhou, Hainan, China
| |
Collapse
|
15
|
Kang BK, Zhu Z, Wang J, Zhou J, Yu S, Zhou X, Zhao Z, Xie A, Lu L, Yang J. Maintenance of adult stem cells from human minor salivary glands via the Wnt signaling pathway. Stem Cell Res Ther 2023; 14:220. [PMID: 37620905 PMCID: PMC10464143 DOI: 10.1186/s13287-023-03445-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Xerostomia is a salivary gland dysfunction that negatively impacts the life quality of patients; however, there is no effective treatment for xerostomia. Bioengineered organs, generated using stem cells obtained from newborn salivary glands and ligated injury models, are a new organ transplantation strategy that could be feasible for xerostomia treatment. Reconstruction of salivary gland organoids by seed cells obtained from human minor salivary glands will offer theoretical fundaments and technology support for clinical application and organ regeneration research. Herein, we aimed to propose a new method for culturing and enriching adult human minor salivary gland stem cells in vitro in a three-dimensional (3D) environment via Wnt signaling activation. METHODS Obtained and characterized human minor salivary gland stem cells (hMSGSCs) with self-organization ability were 3D-cultured to generate organoids. We examined hMSGSCs proliferation and colony formation using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assays. Telomerase reverse transcriptase staining, flow cytometry, immunofluorescence assay, RNA isolation, RT-PCR, and qPCR were performed to assess hMSGSCs structure and the function of reconstructive organoids in vitro. RESULTS hMSGSCs showed typical epithelial-like characteristics, such as positive for CD49f and cell KRT expression. hMSGSCs served as adult stem cells in salivary glands and could differentiate into acinar and duct cells. Upon the addition of Noggin, CHIR99021, and Wnt3A to the 3D culture system, hMSGSCs showed higher LGR5 expression and decreased AMY1B and MUC5B expression. Therefore, the Wnt and bone morphogenetic protein (BMP) pathways are important in regulating hMSGSCs self-organization and differentiation. CONCLUSIONS We showed that the stem cell properties of hMSGSCs in a 3D culture system can be maintained by activating the Wnt signaling pathway and inhibiting the BMP signaling pathway. Our findings contribute new insights on salivary gland organoid generation in vitro.
Collapse
Affiliation(s)
- Bo Kyoung Kang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, China
| | - Zhu Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Shanghai Key Laboratory of Tissue Engineering, Shanghai, 200011, China
| | - Jian Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jia Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shun Yu
- Department of Burns and Plastic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, 214041, China
| | - Xianyu Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhenmin Zhao
- Department of Plastic Surgery, Peking University 3Rd Hospital, NO.49 of North Huayuan Road, Haidian District, Beijing, 100191, China
| | - Aiguo Xie
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Lin Lu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
16
|
Chen W, Liu W, Jiang T, Liu L, He Q, Lin T, Zhang J, Huo L, Xu X, Wang H, Liang D, Liu W. Tongbi Huoluo Decoction alleviates cartilage degeneration in knee osteoarthritis by inhibiting degradation of extracellular matrix. Chin Med 2023; 18:91. [PMID: 37507774 PMCID: PMC10385923 DOI: 10.1186/s13020-023-00802-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is an age-related degenerative disease characterized by abrasion of articular cartilage. Tongbi Huoluo Decoction (TBHLD) has been transformed from the famous traditional Chinese medicine Duhuo Jisheng Decoction, which can effectively alleviate pain symptoms in KOA. However, the active components and mechanisms of TBHLD in treating KOA have not yet been elucidated. The purpose of the study was to demonstrate the molecular mechanism of TBHLD in treating KOA. METHODS The components and targets of TBHLD and KOA were collected from multiple databases, and the protein to protein interaction (PPI) network was constructed. Next, we performed topological calculation and enrichment analysis. Besides, we performed virtual screening for molecular docking and molecular dynamics simulation (MDS). Furthermore, the vitro and vivo experiments were performed to evaluate the validity and mechanism of TBHLD. RESULTS 206 active components and 187 potential targets were screened from Tongbi Huoluo Decoction. A total of 50 intersecting genes were identified between TBHLD and KOA, 20 core targets were calculated by network topology analysis. The core targets were enriched in the ECM interaction pathways. The results of virtual screening for molecular docking and MDS showed that the active components of TBHLD had steady binding conformations with core genes. Moreover, we identified 32 differential serum components in TBHLD-containing serum using LC-MS, including 22 upregulated and 10 downregulated serum components. TBHLD improved the proliferation activity of OA chondrocytes, decreased the expression of Col1a1, Col1a2, Mmp2, Mmp13 in OA chondrocytes, ameliorated the cartilage lesions and restored the cartilage abrasion. CONCLUSION TBHLD inhibited degradation of cartilage ECM by regulating the expression of type I collagens and Mmps to ameliorate cartilage degeneration in KOA.
Collapse
Affiliation(s)
- Weijian Chen
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Weinian Liu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510045, Guangdong, China
| | - Tao Jiang
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Lingyun Liu
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Qi He
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Tianye Lin
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Jiayuan Zhang
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Liwei Huo
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510045, Guangdong, China
| | - Xuemeng Xu
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China
| | - Haibin Wang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China.
| | - Du Liang
- Guangzhou Orthopedic Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510045, Guangdong, China.
| | - Wengang Liu
- The Fifth Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, 510095, China.
- Guangdong Second Hospital of Traditional Chinese Medicine (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China.
| |
Collapse
|
17
|
Wang S, Meng P, Yuan L, Guo X. Analysis of N-glycosylation protein of Kashin-Beck disease chondrocytes derived from induced pluripotent stem cells based on label-free strategies with LC-MS/MS. Mol Omics 2023; 19:454-463. [PMID: 37186116 DOI: 10.1039/d3mo00018d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
We aimed to compare N-glycosylation proteins in Kashin-Beck disease (KBD) chondrocytes and normal chondrocytes derived from induced pluripotent stem cells (iPSCs). KBD and normal iPSCs were reprogrammed from human KBD and normal dermal fibroblasts, respectively. Subsequently, chondrocytes were differentiated from KBD and normal iPSCs separately. Immunofluorescence was utilized to assay the protein markers of iPSCs and chondrocytes. Differential N-glycosylation proteins were screened using label-free strategies with LC-MS/MS. Bioinformatics analyses were utilized to interpret the functions of differential N-glycosylation proteins. Immunofluorescence staining revealed that both KBD-iPSCs and normal-iPSCs strongly expressed pluripotency markers OCT4 and NANOG. Meanwhile, chondrocyte markers collagen II and SOX9 are presented in KBD-iPSC-chondrocytes and normal-iPSC-chondrocytes. We obtained 87 differential N-glycosylation sites which corresponded to 68 differential proteins, which were constructed into 1 cluster. We obtained collagen type I trimer and 9 other biological processes; polysaccharide binding and 9 other molecular functions; regulation of transcription by RNA polymerase II and 9 other cellular components from GO; the Pl3K-Akt signaling pathway and 9 other KEGG pathways; peroxisome and 7 other subcellular locations; and integrin alpha chain, C-terminal cytoplasmic region, conserved site and 9 other classifications of domain annotations, and 2 networks. FGFR3 and LRP1 are expressed at higher levels in KBD-iPSC-chondrocytes, while the expressions of COL2A1, TIMP1, UNC5B, NOG, LEPR, and ITGA1 were down-regulated in KBD-iPSC-chondrocytes. The differential expressions of these N-glycosylation proteins may lead to the abnormal function of KBD chondrocytes.
Collapse
Affiliation(s)
- Sen Wang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, Xi'an, Shaanxi, China.
| | - Peilin Meng
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, Xi'an, Shaanxi, China.
| | - Linlin Yuan
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, Xi'an, Shaanxi, China.
| | - Xiong Guo
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission, Xi'an, Shaanxi, China.
| |
Collapse
|
18
|
Peng Y, Jiang H, Zuo HD. Factors affecting osteogenesis and chondrogenic differentiation of mesenchymal stem cells in osteoarthritis. World J Stem Cells 2023; 15:548-560. [PMID: 37424946 PMCID: PMC10324504 DOI: 10.4252/wjsc.v15.i6.548] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 06/26/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that often involves progressive cartilage degeneration and bone destruction of subchondral bone. At present, clinical treatment is mainly for pain relief, and there are no effective methods to delay the progression of the disease. When this disease progresses to the advanced stage, the only treatment option for most patients is total knee replacement surgery, which causes patients great pain and anxiety. As a type of stem cell, mesenchymal stem cells (MSCs) have multidirectional differentiation potential. The osteogenic differentiation and chondrogenic differentiation of MSCs can play vital roles in the treatment of OA, as they can relieve pain in patients and improve joint function. The differentiation direction of MSCs is accurately controlled by a variety of signaling pathways, so there are many factors that can affect the differentiation direction of MSCs by acting on these signaling pathways. When MSCs are applied to OA treatment, the microenvironment of the joints, injected drugs, scaffold materials, source of MSCs and other factors exert specific impacts on the differentiation direction of MSCs. This review aims to summarize the mechanisms by which these factors influence MSC differentiation to produce better curative effects when MSCs are applied clinically in the future.
Collapse
Affiliation(s)
- Yi Peng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Radiology, Chengdu Xinhua Hospital, Chengdu 610067, Sichuan Province, China
| |
Collapse
|
19
|
Li H, Cao Y, Chang C, Huang W, Su S, Peng Z, Zhang J. Knockdown of circSOD2 ameliorates osteoarthritis progression via the miR-224-5p/PRDX3 axis. J Orthop Surg Res 2023; 18:432. [PMID: 37312219 DOI: 10.1186/s13018-023-03880-9] [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: 09/03/2022] [Accepted: 05/24/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Although the implications of circular RNAs (circRNAs) with the progression of diverse pathological conditions have been reported, the circRNA players in osteoarthritis (OA) are barely studied. METHODS In this study, twenty-five OA patients who received arthroplasty were recruited for cartilage tissue collection. Public circRNA microarray data from Gene Expression Omnibus was retrieved for circRNA identification. An in vitro cell model of OA-related damages was constructed by treating human chondrocytes (CHON-001 cell line) with IL-1β, and circSOD2 siRNA was used to silence circSOD2 expression to study its functional role in apoptosis, inflammatory responses, and extracellular matrix (ECM) degradation. Besides, we investigated the functional interactions among circSOD2, miR-224-5p, and peroxiredoxin 3 (PRDX3) by luciferase reporter assay, RNA-immunoprecipitation assay, and quantitative reverse transcription polymerase chain reaction. RESULTS Our findings revealed the overexpression of circSOD2 in the OA cartilage and cell samples, and circSOD2 knockdown alleviated ECM degradation, inflammation, and apoptosis in CHON-001 cell model. In addition, our findings suggested the regulatory function of circSOD2 knockdown on miR-224-5p expression, while miR-224-5p was capable of downregulating PRDX3 expression. The co-transfection of miR-224-5p inhibitor or pcDNA-PRDX3 could prevent the effect of circSOD2 knockdown. CONCLUSION Hence, our results demonstrated that knockdown of circSOD2 may serve as an intervention strategy to alleviate OA progression through modulating miR-224-5p/PRDX3 signaling axis.
Collapse
Affiliation(s)
- Hao Li
- Department of Sports Medicine, Shenzhen Second People's Hospital, Shenzhen, 518037, Guangdong, China
| | - Yong Cao
- Department of Orthopaedic Emergency, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, Chongqing, 400000, China
| | - Chongfei Chang
- Shenzhen Cheerland Danlun Biomedical Co. Ltd, Shenzhen, 518108, Guangdong, China
| | - Wenping Huang
- Department of Traumatic Orthopedics, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, 9 Jiefang West Road, Yuzhong District, Chongqing, 400000, China
| | - Songchuan Su
- Department of Traumatic Orthopedics, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, 9 Jiefang West Road, Yuzhong District, Chongqing, 400000, China
| | - Zhenggang Peng
- Department of Traumatic Orthopedics, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, 9 Jiefang West Road, Yuzhong District, Chongqing, 400000, China
| | - Jiajin Zhang
- Department of Traumatic Orthopedics, Chongqing Orthopedic Hospital of Traditional Chinese Medicine, 9 Jiefang West Road, Yuzhong District, Chongqing, 400000, China.
| |
Collapse
|
20
|
Motta F, Barone E, Sica A, Selmi C. Inflammaging and Osteoarthritis. Clin Rev Allergy Immunol 2023; 64:222-238. [PMID: 35716253 DOI: 10.1007/s12016-022-08941-1] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 12/15/2022]
Abstract
Osteoarthritis is a highly prevalent disease particularly in subjects over 65 years of age worldwide. While in the past it was considered a mere consequence of cartilage degradation leading to anatomical and functional joint impairment, in recent decades, there has been a more dynamic view with the synovium, the cartilage, and the subchondral bone producing inflammatory mediators which ultimately lead to cartilage damage. Inflammaging is defined as a chronic, sterile, low-grade inflammation state driven by endogenous signals in the absence of infections, occurring with aging. This chronic status is linked to the production of reactive oxygen species and molecules involved in the development of age-related disease such as cancer, diabetes, and cardiovascular and neurodegenerative diseases. Inflammaging contributes to osteoarthritis development where both the innate and the adaptive immune response are involved. Elevated systemic and local inflammatory cytokines and senescent molecules promote cartilage degradation, and antigens derived from damaged joints further trigger inflammation through inflammasome activation. B and T lymphocyte populations also change with inflammaging and OA, with reduced regulatory functions, thus implicating self-reactivity as an additional mechanism of joint damage. The discovery of the underlying pathogenic pathways may help to identify potential therapeutic targets for the management or the prevention of osteoarthritis. We will provide a comprehensive evaluation of the current literature on the role of inflammaging in osteoarthritis and discuss the emerging therapeutic strategies.
Collapse
Affiliation(s)
- Francesca Motta
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy
| | - Elisa Barone
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy
| | - Antonio Sica
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, Milan, Italy.,Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", Largo Donegani 2, 28100, Novara, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089, Rozzano, Milan, Italy. .,Department of Biomedical Sciences, Humanitas University, via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy.
| |
Collapse
|
21
|
Yu L, Luo R, Qin G, Zhang Q, Liang W. Efficacy and safety of anti-interleukin-1 therapeutics in the treatment of knee osteoarthritis: a systematic review and meta-analysis of randomized controlled trials. J Orthop Surg Res 2023; 18:100. [PMID: 36782214 PMCID: PMC9923921 DOI: 10.1186/s13018-023-03590-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
OBJECTIVE We aimed to evaluate the efficacy and safety of anti-interleukin-1 therapeutics, including IL-1 antibodies, interleukin-1 receptor antagonists (IL-1 Ras) and IL-1 inhibitors, for knee osteoarthritis (KOA) treatment. METHODS Databases (Medline, Embase, Web of Science and CENTRAL) and ClinicalTrials.gov were systematically searched for randomized controlled trials (RCTs) of anti-interleukin-1 therapeutics from inception to August 31, 2022. The outcomes were the mean change in pain and function scores and the risk of adverse effects (AEs). RESULTS In the 12 studies included, anti-interleukin-1 therapeutics were superior to placebo in terms of pain relief (standardized mean difference [SMD] = - 0.38, 95% confidence interval [CI] = - 1.82 to - 0.40, p < 0.001, I2 = 77%) and functional improvement (SMD = - 1.11, 95% CI = - 1.82 to - 0.40, p = 0.002, I2 = 96%). The incidence of any AE (risk ratio [RR] = 1.02, 95% CI = 0.88-1.18, p < 0.001, I2 = 76%) was higher following treatment with anti-interleukin-1 therapeutics than placebo, while no significant difference was found in the incidence of serious AEs (SAEs) or discontinuations due to AEs. Subgroup analyses showed that IL-1 antibodies and the IL-1 inhibitor provided pain relief (IL-1 antibodies: SMD = - 0.61, 95% CI = - 0.92 to - 0.31, p < 0.001; IL-1 inhibitor: SMD = - 0.39, 95% CI = - 0.72 to - 0.06, p = 0.02, I2 = 74.0%) and functional improvement (IL-1 antibodies: SMD = - 1.75, 95% CI = - 2.10 to - 1.40, p < 0.001; IL-1 inhibitor: SMD = - 0.28, 95% CI = - 0.83 to 0.27, p = 0.31, I2 = 88%) superior to those of placebo, whereas IL-1 Ras did not. However, the IL-1 inhibitor increased the incidence of any AE (RR = 1.35, 95% CI = 0.92-1.98, p < 0.001, I2 = 85%) but not the risk of SAEs or discontinuations due to AEs. IL-1 antibodies and IL-1 Ras showed no difference in safety compared with placebo. CONCLUSIONS Anti-interleukin-1 therapeutics could relieve OA-related pain and improve function, but is probably associated with an increased risk of adverse events. Specially, IL-1 antibodies and an IL-1 inhibitor could relieve OA-related pain and improve function, whereas IL-1 Ras could not. IL-1 antibodies and IL-1 Ras were relatively safe options, but IL-1 inhibitors were associated with safety concerns.
Collapse
Affiliation(s)
- Lizhi Yu
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, 124 Yuejin Road, Liuzhou, 545001, Guangxi Province, China
| | - Raoshan Luo
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, 124 Yuejin Road, Liuzhou, 545001, Guangxi Province, China
| | - Gang Qin
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, 124 Yuejin Road, Liuzhou, 545001, Guangxi Province, China
| | - Qinyan Zhang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, 124 Yuejin Road, Liuzhou, 545001, Guangxi Province, China
| | - Weiming Liang
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, 124 Yuejin Road, Liuzhou, 545001, Guangxi Province, China.
| |
Collapse
|
22
|
Yao Q, Wu X, Tao C, Gong W, Chen M, Qu M, Zhong Y, He T, Chen S, Xiao G. Osteoarthritis: pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther 2023; 8:56. [PMID: 36737426 PMCID: PMC9898571 DOI: 10.1038/s41392-023-01330-w] [Citation(s) in RCA: 346] [Impact Index Per Article: 173.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/06/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder that leads to disability and affects more than 500 million population worldwide. OA was believed to be caused by the wearing and tearing of articular cartilage, but it is now more commonly referred to as a chronic whole-joint disorder that is initiated with biochemical and cellular alterations in the synovial joint tissues, which leads to the histological and structural changes of the joint and ends up with the whole tissue dysfunction. Currently, there is no cure for OA, partly due to a lack of comprehensive understanding of the pathological mechanism of the initiation and progression of the disease. Therefore, a better understanding of pathological signaling pathways and key molecules involved in OA pathogenesis is crucial for therapeutic target design and drug development. In this review, we first summarize the epidemiology of OA, including its prevalence, incidence and burdens, and OA risk factors. We then focus on the roles and regulation of the pathological signaling pathways, such as Wnt/β-catenin, NF-κB, focal adhesion, HIFs, TGFβ/ΒΜP and FGF signaling pathways, and key regulators AMPK, mTOR, and RUNX2 in the onset and development of OA. In addition, the roles of factors associated with OA, including MMPs, ADAMTS/ADAMs, and PRG4, are discussed in detail. Finally, we provide updates on the current clinical therapies and clinical trials of biological treatments and drugs for OA. Research advances in basic knowledge of articular cartilage biology and OA pathogenesis will have a significant impact and translational value in developing OA therapeutic strategies.
Collapse
Affiliation(s)
- Qing Yao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Chu Tao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Weiyuan Gong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Mingjue Chen
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Minghao Qu
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yiming Zhong
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tailin He
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
23
|
Effects of Tuina on cartilage degradation and chondrocyte terminal differentiation in rats with knee osteoarthritis (KOA) via the Wnt/β-catenin signaling pathway. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2023. [DOI: 10.1007/s11726-023-1354-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
24
|
Heng BC, Bai Y, Li X, Lim LW, Li W, Ge Z, Zhang X, Deng X. Electroactive Biomaterials for Facilitating Bone Defect Repair under Pathological Conditions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204502. [PMID: 36453574 PMCID: PMC9839869 DOI: 10.1002/advs.202204502] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/24/2022] [Indexed: 06/02/2023]
Abstract
Bone degeneration associated with various diseases is increasing due to rapid aging, sedentary lifestyles, and unhealthy diets. Living bone tissue has bioelectric properties critical to bone remodeling, and bone degeneration under various pathological conditions results in significant changes to these bioelectric properties. There is growing interest in utilizing biomimetic electroactive biomaterials that recapitulate the natural electrophysiological microenvironment of healthy bone tissue to promote bone repair. This review first summarizes the etiology of degenerative bone conditions associated with various diseases such as type II diabetes, osteoporosis, periodontitis, osteoarthritis, rheumatoid arthritis, osteomyelitis, and metastatic osteolysis. Next, the diverse array of natural and synthetic electroactive biomaterials with therapeutic potential are discussed. Putative mechanistic pathways by which electroactive biomaterials can mitigate bone degeneration are critically examined, including the enhancement of osteogenesis and angiogenesis, suppression of inflammation and osteoclastogenesis, as well as their anti-bacterial effects. Finally, the limited research on utilization of electroactive biomaterials in the treatment of bone degeneration associated with the aforementioned diseases are examined. Previous studies have mostly focused on using electroactive biomaterials to treat bone traumatic injuries. It is hoped that this review will encourage more research efforts on the use of electroactive biomaterials for treating degenerative bone conditions.
Collapse
Affiliation(s)
- Boon Chin Heng
- Central LaboratoryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- School of Medical and Life SciencesSunway UniversityDarul EhsanSelangor47500Malaysia
| | - Yunyang Bai
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xiaochan Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Lee Wei Lim
- Neuromodulation LaboratorySchool of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong KongP. R. China
| | - Wang Li
- Department of Biomedical EngineeringPeking UniversityBeijing100871P. R. China
| | - Zigang Ge
- Department of Biomedical EngineeringPeking UniversityBeijing100871P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of Dental Materials & Dental Medical Devices Testing CenterPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Engineering Research Center of Oral Biomaterials and Digital Medical DevicesNMPA Key Laboratory for Dental MaterialsBeijing Laboratory of Biomedical Materials & Beijing Key Laboratory of Digital StomatologyPeking University School and Hospital of StomatologyBeijing100081P. R. China
| |
Collapse
|
25
|
Tong Z, Yang X, Li J. Research progress on the mechanism of interleukin-1β on epiphyseal plate chondrocytes. Eur J Med Res 2022; 27:313. [PMID: 36575508 PMCID: PMC9793524 DOI: 10.1186/s40001-022-00893-8] [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: 09/11/2022] [Accepted: 11/08/2022] [Indexed: 12/28/2022] Open
Abstract
Epiphyseal plate injury, a common problem in pediatric orthopedics, may result in poor bone repair or growth defects. Epiphyseal plate, also known as growth plate is a layer of hyaline cartilage tissue between the epiphysis and metaphyseal and has the ability to grow longitudinally. Under normal physiological conditions, the epiphyseal plate has a certain axial resistance to stress, but it is fragile in growth phase and can be damaged by excessive stress, leading to detachment or avulsion of the epiphysis, resulting in life-long devastating consequences for patients. There is an obvious inflammatory response in the phase of growth plate injury, the limited physiological inflammatory response locally favors tissue repair and the organism, but uncontrolled chronic inflammation always leads to tissue destruction and disease progression. Interleukin-1β (IL-1β), as representative inflammatory factors, not only affect the inflammatory phase response to bone and soft tissue injury, but have a potentially important role in the later repair phase, though the exact mechanism is not fully understood. At present, epiphyseal plate injuries are mainly treated by corrective and reconstructive surgery, which is highly invasive with limited effectiveness, thus new therapeutic approaches are urgently needed, so a deeper understanding and exploration of the pathological mechanisms of epiphyseal plate injuries at the cellular molecular level is an entry point. In this review, we fully introduced the key role of IL-1 in the progression of epiphyseal plate injury and repair, deeply explored the mechanism of IL-1 on the molecular transcript level and endocrine metabolism of chondrocytes from multiple aspects, and summarized other possible mechanisms to provide theoretical basis for the clinical treatment and in-depth study of epiphyseal plate injury in children.
Collapse
Affiliation(s)
- Ziyuan Tong
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 114000 Liaoning China
| | - Xu Yang
- grid.412467.20000 0004 1806 3501Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 114000 Liaoning China
| | - Jianjun Li
- grid.412467.20000 0004 1806 3501Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, 114000 Liaoning China ,grid.412467.20000 0004 1806 3501Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 114000 Liaoning China
| |
Collapse
|
26
|
Li C, Li W, Pu G, Wu J, Qin F. Exosomes derived from miR-338-3p-modified adipose stem cells inhibited inflammation injury of chondrocytes via targeting RUNX2 in osteoarthritis. J Orthop Surg Res 2022; 17:567. [PMID: 36572886 PMCID: PMC9791748 DOI: 10.1186/s13018-022-03437-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease that is one of the main causes of disability in middle-aged and elderly people. Adipose stem cell (ASC)-derived exosomes (ASC-Exo) could repair cartilage damage and treat OA. MiRNA-338-3p expression was confirmed to play a role in inhibiting proinflammatory cytokines. Herein, we aimed to explore the mechanism by which exosomes derived from miR-338-3p overexpressing ASCs protects chondrocytes from interleukin (IL)-1β-induced chondrocyte change. METHODS Exosomes were extracted from ASCs transfected with miR-338-3p or its antisense inhibitor. The ASC-Exos (miR-338-3p silencing/overexpression) were incubated with IL-1β-induced ATDC5 cells, followed by evaluation of the chondrocyte proliferation, degradation, and inflammation injury. RESULTS In vitro results revealed that ASC-Exos inhibited the expression of prostaglandin E2 (PGE2), IL-6, IL-1β, and TNF-α, as well as promoted the proliferation of ATDC5 cells. Moreover, ASC-Exos inhibited inflammation injury and degradation of ATDC5 cells by transferring miR-338-3p. Luciferase reporter assays showed that RUNX2 was a target gene of miR-338-3p. Additionally, RUNX2 overexpression in ATDC5 cells reversed the protective effect of miR-338-3p on chondrocytes. Taken together, this study demonstrated that exosomes secreted from miR-338-3p-modified ASCs were effective in the repair of IL-1β-induced chondrocyte change by inhibiting RUNX2 expression. CONCLUSIONS Our result provided valuable data for understanding the mechanism of ASC-Exos in OA treatment.
Collapse
Affiliation(s)
- ChunLiang Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Wei Li
- grid.469564.cDepartment of Orthopedic, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - GengZang Pu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - JingWen Wu
- grid.469564.cDepartment of Emergency Surgery, Qinghai Provincial People’s Hospital, Xining, 810006 Qinghai China
| | - Feng Qin
- grid.459333.bDepartment of Endocrinology, Qinghai University Affiliated Hospital, Chengxi District, No. 6, Xichuan South Road, Xining, 810006 Qinghai China
| |
Collapse
|
27
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
28
|
Liu H, Wang C, Sun X, Zhan C, Li Z, Qiu L, Luo R, Liu H, Sun X, Li R, Zhang J. Silk Fibroin/Collagen/Hydroxyapatite Scaffolds Obtained by 3D Printing Technology and Loaded with Recombinant Human Erythropoietin in the Reconstruction of Alveolar Bone Defects. ACS Biomater Sci Eng 2022; 8:5245-5256. [PMID: 36336837 DOI: 10.1021/acsbiomaterials.2c00690] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The fast osteogenesis of the large alveolar fossa and the maintenance of the height of the alveolar ridge after tooth extraction have always been a clinical challenge. Therefore, this work describes the creation of innovative silk fibroin/collagen/hydroxyapatite (SCH) biological scaffolds by 3D printing technology, which are loaded with recombinant human erythropoietin (rh-EPO) for the reconstruction of bone defects. Low-temperature 3D printing can maintain the biological activity of silk fibroin and collagen. The SCH scaffolds showed the ideal water absorption and porosity, being a sustained-release carrier of rh-EPO. The optimized scaffolds had ideal mechanical properties in vitro, and MC3T3-E1 cells could easily adhere and proliferate on it. In vivo experiments in rabbits demonstrated that the composite scaffolds gradually degraded and promoted the accumulation and proliferation of osteoblasts and the formation of collagen fibers, significantly promoting the reconstruction of mandibular defects. In this study, a novel composite biological scaffold was prepared using 3D printing technology, and the scaffold was innovatively combined with the multifunctional growth factor rh-EPO. This provides a new optimized composite material for the reconstruction of irregular mandible defects, and this biomaterial is promising for clinical reconstruction of alveolar bone defects.
Collapse
Affiliation(s)
- Han Liu
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Chao Wang
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Xiaoqian Sun
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Chaojun Zhan
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Zixiao Li
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Lin Qiu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100034, China
| | - Rui Luo
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China.,School of Medicine, Nankai University, Tianjin 300071, China
| | - Hao Liu
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Xiaodi Sun
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Ruixin Li
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Jun Zhang
- Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| |
Collapse
|
29
|
Guowei G, Yuzhong Z, Xuan Z, Zhi D, Juanhui D, Jing W, Peikui Y, Xiangzhi L, Zhen W. Zhuanggu Guanjie herbal formula mitigates osteoarthritis via the NF-κB transduction mechanism. Front Pharmacol 2022; 13:896397. [PMID: 36532734 PMCID: PMC9751418 DOI: 10.3389/fphar.2022.896397] [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: 03/15/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
The Zhuanggu Guanjie herbal formula has been a famous Chinese prescription for treating bone diseases since time immemorial. The anti-osteoarthritis (OA) properties of this botanical prescription are well documented in the Chinese Pharmacopoeia. However, the detailed mechanisms behind the phenomenon have not been elucidated. Hence, we aimed to investigate the anti-OA efficacy of the Zhuanggu Guanjie herbal formula and its underlying mechanism. The anti-OA properties of Zhuanggu Guanjie capsule (ZGC) were determined by the cytokine contents and inflammatory-related proteins, which were measured by RT-PCR, flow cytometry, Western blot, and laser confocal assay in ATDC5 cells. The levels of interleukin-6, tumor necrosis factor-α, inducible nitric oxide synthase, cyclooxygenase-2, and prostaglandin synthesis E2 have been markedly reduced after being treated with ZGC for 48 h in a dose-dependent manner. Furthermore, ZGC prevented the translocation of NF-κB from the cytosol to the nucleus. On the other hand, we used the mono-iodoacetate (MIA)-induced OA model to confirm the in vivo efficacies of this herbal formula. Oral administration of ZGC attenuated MIA-induced OA damage through changes in histopathological and knee joint volumes. The serum matrix metalloproteinase-13 contents in the ZGC treatment group declined as compared to those in the MIA model group. Through our in vitro and in vivo studies, we confirmed the anti-OA efficacy of ZGC and uncovered its detailed mechanism, and this treatment shed light on OA pathophysiology.
Collapse
Affiliation(s)
- Gong Guowei
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, China,Guangdong Provincial Key Laboratory of Functional Substances in Medicinal Edible Resources and Healthcare Products, Hanshan Normal University, Chaozhou, China,*Correspondence: Gong Guowei, ; Zheng Yuzhong,
| | - Zheng Yuzhong
- Guangdong Provincial Key Laboratory of Functional Substances in Medicinal Edible Resources and Healthcare Products, Hanshan Normal University, Chaozhou, China,*Correspondence: Gong Guowei, ; Zheng Yuzhong,
| | - Zhou Xuan
- Department of Bioengineering, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Dai Zhi
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| | - Duan Juanhui
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| | - Wang Jing
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen, China
| | - Yang Peikui
- Department of Medical Laboratory, Chaozhou People’s Hospital, Chaozhou, China
| | - Liu Xiangzhi
- Department of Medical Laboratory, Chaozhou People’s Hospital, Chaozhou, China
| | - Wen Zhen
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
30
|
Circ_0022383 alleviates IL-1β-induced apoptosis, inflammation and extracellular matrix degeneration in osteoarthritis cell model by miR-3619-5p/SIRT1 axis. Int Immunopharmacol 2022; 112:109289. [DOI: 10.1016/j.intimp.2022.109289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022]
|
31
|
Ivanenko KA, Prassolov VS, Khabusheva ER. Transcription Factor Sp1 in the Expression of Genes Encoding Components of Mapk, JAK/STAT, and PI3K/Akt Signaling Pathways. Mol Biol 2022. [DOI: 10.1134/s0026893322050089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
32
|
Hu Y, Cui J, Liu H, Wang S, Zhou Q, Zhang H, Guo J, Cao L, Chen X, Xu K, Su J. Single-cell RNA-sequencing analysis reveals the molecular mechanism of subchondral bone cell heterogeneity in the development of osteoarthritis. RMD Open 2022. [PMCID: PMC9462384 DOI: 10.1136/rmdopen-2022-002314] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The cellular composition and underlying spatiotemporal transformation processes of subchondral bone in osteoarthritis (OA) remain unknown. Herein, various cell subsets from tibial plateau of patients with OA are identified, and the mechanism of subchondral microstructure alteration is elaborated using single-cell RNA sequencing technique. We identified two novel endothelial cell (EC) populations characterised by either exosome synthesis and inflammation response or vascular function and angiogenesis. Three osteoblast (OB) subtypes are introduced, separately related to vascularisation, matrix manufacturing and matrix mineralisation. The distinct roles and functions of these novel phenotypes in OA development are further discussed as well as interaction network between these subpopulations. The variation tendency of each population is testified in a destabilisation of the medial meniscus mouse model. The identification of cell types demonstrates a novel taxonomy and mechanism for ECs and OBs inside subchondral bone area provides new insights into the physiological and pathological behaviours of subchondral bone in OA pathogenesis.
Collapse
Affiliation(s)
- Yan Hu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Jin Cui
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Qirong Zhou
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Hao Zhang
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Jiawei Guo
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Baoshan Luodian Hospital, Shanghai, China
| | - Xiao Chen
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedics, Changhai Hospital, Shanghai, China
| |
Collapse
|
33
|
Zhang Y, Zhao P, Li S, Mu X, Wang H. CircSCAPER knockdown attenuates IL-1β-induced chondrocyte injury by miR-127-5p/TLR4 axis in osteoarthritis. Autoimmunity 2022; 55:577-586. [PMID: 35993243 DOI: 10.1080/08916934.2022.2103798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
PURPOSE Osteoarthritis (OA) is a chronic inflammatory degenerative disease characterized by articular cartilage degradation. Circular RNAs have been shown to play significant roles in OA process. Herein, this work aimed to investigate the potential role and mechanism of circSCAPER in OA progression. METHODS Levels of circSCAPER, miR-127-5p and toll-like receptor 4 (TLR4) were detected by qRT-PCR or western blotting. Cell apoptosis was determined by flow cytometry. The expression of Aggrecan and Matrix metallopeptidase was examined using western blot to assess extracellular matrix (ECM) degradation. Inflammatory response and oxidative stress were determined by measuring the release of inflammatory factors, along with the generation of intracellular reactive oxygen species and malondialdehyde. The interaction between miR-127-5p and circSCAPER or TLR4 was determined by dual-luciferase reporter, RNA immunoprecipitation and pull-down assays. RESULTS Chondrocytes were treated with interleukin-1β (IL-1β) to mimic OA condition in vitro. CircSCAPER was increased in OA cartilages and IL-1β-induced chondrocytes. Functionally, knockdown of circSCAPER attenuated IL-1β-evoked apoptosis, ECM degradation, inflammation and oxidative stress in vitro. CircSCAPER up-regulation in OA cartilages was discovered to be accompanied by decreased miR-127-5p and increased TLR4. Mechanistically, circSCAPER acted as a sponge for miR-127-5p to positively regulate TLR4 expression in chondrocytes. IL-1β treatment reduced miR-127-5p expression but up-regulated TLR4 expression, re-expression of miR-127-5p suppressed IL-1β-caused chondrocyte injury, which was abolished by TLR4 overexpression. Moreover, miR-127-5p inhibition reversed the protective action of circSCAPER knockdown on chondrocytes under IL-1β treatment. CONCLUSION CircSCAPER silencing protected against IL-1β-induced apoptosis, ECM degradation, inflammation and oxidative stress in chondrocytes via miR-127-5p/TLR4 axis.
Collapse
Affiliation(s)
- Yuchang Zhang
- Department of Orthopedics, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Ping Zhao
- Department of Orthopedics, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Sen Li
- Department of Orthopedics, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xiangqian Mu
- Department of Orthopedics, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Huaqi Wang
- Department of Orthopedics, Ankang Traditional Chinese Medicine Hospital, Ankang, China
| |
Collapse
|
34
|
Terzi MY, Okuyan HM, Karaboğa İ, Gökdemir CE, Tap D, Kalacı A. Urotensin-II Prevents Cartilage Degeneration in a Monosodium Iodoacetate-Induced Rat Model of Osteoarthritis. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10448-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Zhang RZ, Shi Q, Zhao H, Pan GQ, Shao LH, Wang JF, Liu HW. In vivo study of dual functionalized mussel-derived bioactive peptides promoting 3D-printed porous Ti6Al4V scaffolds for repair of rabbit femoral defects. J Biomater Appl 2022; 37:942-958. [PMID: 35856165 DOI: 10.1177/08853282221117209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The 3D printed porous titanium alloy scaffolds are beneficial to enhance angiogenesis, osteoblast adhesion, and promote osseointegration. However, titanium alloys are biologically inert, which makes the bond between the implant and bone tissue weak and prone to loosening. Inspired by the natural biological marine mussels, we designed four-claw-shaped mussel-derived bioactive peptides for the decoration of porous titanium alloy scaffolds: adhesion peptide-DOPA, anchoring peptide-RGD and osteogenic-inducing peptide-BMP-2. And the bifunctionalization of 3D-printed porous titanium alloy scaffolds was evaluated in vivo in a rabbit model of bone defect with excellent promotion of osseointegration and mechanical stability. Our results show that the in vivo osseointegration ability of the modified 3D printed porous titanium alloy test piece is significantly improved, and the bifunctional polypeptide coating group E has the strongest osseointegration ability. In conclusion, our experimental design partially solves the problems of stress shielding effect and biological inertness, and provides a convenient and feasible method for the clinical application of titanium alloy implants in biomedical implant materials.
Collapse
Affiliation(s)
| | - Qin Shi
- 12582Suzhou University, Suzhou, China
| | - Huan Zhao
- 12582Suzhou University, Suzhou, China
| | | | | | | | - Hong Wei Liu
- 599923Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
36
|
Wang L, Zhao L, Shen L, Fang Q, Yang Z, Wang R, Wu Q, Xie Y. Comparison of the effects of autologous and allogeneic purified platelet-rich plasma on cartilage damage in a rabbit model of knee osteoarthritis. Front Surg 2022; 9:911468. [PMID: 35910465 PMCID: PMC9334772 DOI: 10.3389/fsurg.2022.911468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Background Purified platelet-rich plasma (P-PRP) is gradually being used in the treatment of osteoarthritis (OA), and its sources are mainly divided into autologous and allogeneic blood. However, it is unclear whether autologous PRP is more effective or allogeneic PRP is superior. Objective In this study, autologous and allogeneic P-PRP was injected at early stage of KOA in rabbits, and then the differences in the efficacy of the two P-PRPs against KOA were compared from several perspectives, including pathological histology and immunohistochemistry. Method Experimental rabbits were divided into normal group (n = 8), model group (n = 8), autologous P-PRP group (n = 8), and allogeneic P-PRP group (n = 8) using a random number table method. The normal and model groups did not receive any treatment, and the autologous P-PRP and allogeneic P-PRP groups received intra-articular injections of autologous and allogeneic P-PRP, respectively, to observe the changes in the gross specimens of the knee joints of the experimental rabbits in each group. The histopathological changes of chondrocytes were also observed by HE-stained sections of articular cartilage, and the expression of chondrocytes Bone morphogenetic protein-2 (BMP-2) and Sox9 were detected by immunohistochemistry. Results Compared with the allogeneic P-PRP group, the differences were statistically significant (P < 0.05) in the gross specimens and pathological histological findings in the autologous PRP group. Immunohistochemical results showed that the expression of BMP-2 and Sox9 was elevated in both the autologous P-PRP group and the allogeneic P-PRP group compared with the model group, and the expression of BMP-2 was higher in the autologous P-PRP group than in the allogeneic P-PRP group, with a statistically significant difference (P < 0.05), while there was no difference in the expression of Sox9 between the two groups (P > 0.05). Conclusion Intra-articular injection of autologous P-PRP activated the expression of BMP-2 and Sox9 in chondrocytes and effectively improved KOA cartilage repair and reduced bone redundancy and joint fluid formation, and its efficacy was superior to that of intra-articular injection of allogeneic P-PRP.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Luting Zhao
- Department of Rehabilitation Medicine, The First People’s Hospital of Ziyang, Ziyang, China
| | - Lianwei Shen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
- Department of Rehabilitation Medicine, Shandong University Cheeloo College of Medicine, Jinan, China; Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qilin Fang
- Department of Rehabilitation Medicine, The First People’s Hospital of Ziyang, Ziyang, China
| | - Zhenglei Yang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Rongrong Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qing Wu
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Correspondence: Qing Wu Yulei Xie
| | - Yulei Xie
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Faculty of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Correspondence: Qing Wu Yulei Xie
| |
Collapse
|
37
|
Chang SLY, Lin YY, Liu SC, Tsai YS, Lin SW, Chen YL, Chen CC, Ko CY, Chen HT, Chen WC, Tang CH. Oral Administration of Clostridium butyricum GKB7 Ameliorates Signs of Osteoarthritis in Rats. Cells 2022; 11:2169. [PMID: 35883610 PMCID: PMC9323988 DOI: 10.3390/cells11142169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative and painful inflammatory joint disease affecting the cartilage, bone, and synovial membranes, without any effective treatment that targets the underlying mechanisms of OA. Our study evaluated the therapeutic effects of a live probiotic strain, Clostridium butyricum GKB7, administered for 6 weeks to rats with knee OA (KOA) induced by anterior cruciate ligament transection (ACLT) of the right knee. All rats underwent weekly weight-bearing behavioral testing and body weight measurements. At 6 weeks, all rats were sacrificed, and the right hind knees were collected for micro-computed tomography imaging and histopathological and immunohistochemical analyses. Compared with rats in the ACLT-only group, ACLT rats administered the probiotic exhibited dramatic improvements in pain-related behavior from postoperative week 2, had significantly less osseous and cartilaginous damage at week 6, and significantly lower levels of the inflammatory markers interleukin 1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) in cartilage and synovium sections. C. butyricum GKB7 appeared to slow or even reverse OA progression and is worth investigating as a novel therapeutic for OA.
Collapse
Affiliation(s)
- Sunny Li-Yun Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (S.L.-Y.C.); (C.-Y.K.)
- School of Medicine, China Medical University, Taichung 404333, Taiwan;
| | - Yen-You Lin
- School of Medicine, China Medical University, Taichung 404333, Taiwan;
| | - Shan-Chi Liu
- Department of Medical Education and Research, Beigang Hospital, China Medical University, Yunlin 651012, Taiwan;
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (Y.-S.T.); (S.-W.L.); (Y.-L.C.)
| | - Shih-Wei Lin
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (Y.-S.T.); (S.-W.L.); (Y.-L.C.)
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan 325002, Taiwan; (Y.-S.T.); (S.-W.L.); (Y.-L.C.)
| | - Chin-Chu Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei 106617, Taiwan;
- Department of Food Science, Nutrition and Nutraceutical Biotechnology, Shih Chien University, Taipei 104036, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Chih-Yuan Ko
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (S.L.-Y.C.); (C.-Y.K.)
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 404333, Taiwan;
| | - Hsien-Te Chen
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung 404333, Taiwan;
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung 404333, Taiwan
| | - Wei-Cheng Chen
- Department of Medicine, MacKay Medical College, New Taipei 25245, Taiwan
- Division of Sports Medicine & Surgery, Department of Orthopedic Surgery, MacKay Memorial Hospital, Taipei 104217, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (S.L.-Y.C.); (C.-Y.K.)
- School of Medicine, China Medical University, Taichung 404333, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 404333, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung 40354, Taiwan
| |
Collapse
|
38
|
Jiawei Yanghe Decoction Regulates Bone-Lipid Balance through the BMP-SMAD Signaling Pathway to Promote Osteogenic Differentiation of Bone Mesenchymal Stem Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2885419. [PMID: 35769158 PMCID: PMC9236768 DOI: 10.1155/2022/2885419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 05/21/2022] [Indexed: 11/17/2022]
Abstract
Background The Jiawei Yanghe decoction (JWYHD) is a traditional Chinese medicine formula for the treatment of osteoporosis, but its therapeutic mechanism has not been fully elucidated, and the therapeutic target of the intervention disease needs to be further verified. The dysfunction of bone mesenchymal stem cells (BMSCs) is considered to be an important pathogenesis of postmenopausal osteoporosis (PMOP). The purpose of this study was to explore how JWYHD regulates BMSC differentiation through the BMP-SMAD signal pathway. Methods In the in vivo study, we used an ovariectomized PMOP rat (n = 36, 2-month-old, 200 ± 20 g) model and femur micro-CT analysis to study the effect of JWYHD on bone loss in rats. By immunofluorescence, the translocation expression of BMP2, a key protein in the pathway, was detected. Serum bone metabolism was detected by an enzyme-linked immunosorbent assay (ELISA). Alkaline phosphatase (ALP) activity was detected by alkaline phosphatase staining (ALPS), osteogenesis and matrix mineralization were detected by alizarin red staining (ARS), the adipogenic ability of BMSCs was detected by oil red staining (ORS), and CFU is used to detect the ability of cells to form colonies. The expression of related proteins was detected by western blotting. Results In vivo and in vitro, the OP phenotypes of SD rats induced by ovariectomy (OVX) included impaired bone mineral density and microstructure, abnormal bone metabolism, and impaired MSC differentiation potential. JWYHD treatment reversed this trend and restored the differentiation potential of MSCs. JWYHD medicated serum and direct intervention of drugs activated the BMP-SMAD signaling pathway, promoted the osteogenic differentiation of BMSCs, and inhibited their adipogenic differentiation. Conclusions Our data identified that JWYHD is an effective alternative drug for the treatment of PMOP that functions to stimulate the differentiation of BMSCs into osteoblasts in the BMP-SMAD signaling-dependent mechanism.
Collapse
|
39
|
Xu T, Yang D, Liu K, Gao Q, Liu Z, Li G. Miya Improves Osteoarthritis Characteristics via the Gut-Muscle-Joint Axis According to Multi-Omics Analyses. Front Pharmacol 2022; 13:816891. [PMID: 35668932 PMCID: PMC9163738 DOI: 10.3389/fphar.2022.816891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The gut microbiota is associated with osteoarthritis (OA) progression. Miya (MY) is a product made from Clostridium butyricum, a member of gut microbiota. This study was conducted to investigate the effects of MY on OA and its underlying mechanisms. Methods: An OA rat model was established, and MY was used to treat the rats for 4 weeks. Knee joint samples from the rats were stained with hematoxylin-eosin, and fecal samples from the OA and OA+MY groups were subjected to 16S rDNA sequencing and metabolomic analysis. The contents of succinate dehydrogenase and muscle glycogen in the tibia muscle were determined, and related genes and proteins were detected using quantitative reverse transcription polymerase chain reaction and western blotting. Results: Hematoxylin and eosin staining showed that treatment with MY alleviated the symptoms of OA. According to the sequencing results, MY significantly increased the Chao1, Shannon, and Pielou evenness values compared to those in the untreated group. At the genus level, the abundances of Prevotella, Ruminococcus, Desulfovibrio, Shigella, Helicobacter, and Streptococcus were higher in the OA group, whereas Lactobacillus, Oscillospira, Clostridium, and Coprococcus were enriched after MY treatment. Metabolomic analysis revealed 395 differentially expressed metabolites. Additionally, MY treatment significantly increased the succinate dehydrogenase and muscle glycogen contents in the muscle caused by OA (p > 0.05). Finally, AMPK, Tfam, Myod, Ldh, Chrna1, Chrnd, Rapsyn, and Agrin were significantly downregulated in the muscles of OA mice, whereas Lcad, Mcad, and IL-1β were upregulated; MY significantly reversed these trends induced by OA. Conclusions: MY may promote the repair of joint damage and protect against OA via the gut-muscle-joint axis.
Collapse
Affiliation(s)
- Tianyang Xu
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong Yang
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kaiyuan Liu
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qiuming Gao
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhongchen Liu
- Department of General Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Zhongchen Liu, ; Guodong Li,
| | - Guodong Li
- Department of Orthopedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Zhongchen Liu, ; Guodong Li,
| |
Collapse
|
40
|
Hong Y, Shan S, Gu Y, Huang H, Zhang Q, Han Y, Dong Y, Liu Z, Huang M, Ren T. Malfunction of airway basal stem cells plays a crucial role in pathophysiology of tracheobronchopathia osteoplastica. Nat Commun 2022; 13:1309. [PMID: 35288560 PMCID: PMC8921516 DOI: 10.1038/s41467-022-28903-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/15/2022] [Indexed: 11/25/2022] Open
Abstract
Understanding disease-associated stem cell abnormality has major clinical implications for prevention and treatment of human disorders, as well as for regenerative medicine. Here we report a multifaceted study on airway epithelial stem cells in Tracheobronchopathia Osteochondroplastica (TO), an under-detected tracheobronchial disorder of unknown etiology and lack of specific treatment. Epithelial squamous metaplasia and heterotopic bone formation with abnormal cartilage proliferation and calcium deposits are key pathological hallmarks of this disorder, but it is unknown whether they are coincident or share certain pathogenic mechanisms in common. By functional evaluation and genome-wide profiling at both transcriptional and epigenetic levels, we reveal a role of airway basal cells in TO progression by acting as a repository of inflammatory and TGFβ-BMP signals, which contributes to both epithelial metaplasia and mesenchymal osteo-chondrogenesis via extracellular signaling and matrix remodeling. Restoration of microenvironment by cell correction or local pathway intervention may provide therapeutic benefits. Tracheobronchopathia osteoplastica (TO), is an underreported affliction characterized by squamous metaplasia and heterotopic bone formation in trachea and bronchi. Here the authors apply functional, as well as genome-wide transcriptional and epigenetic profiling to identify airway basal cells dysfunction underlying TO.
Collapse
|
41
|
Pan X, Yuan S, Xun X, Fan Z, Xue X, Zhang C, Wang J, Deng J. Long-Term Recruitment of Endogenous M2 Macrophages by Platelet Lysate-Rich Plasma Macroporous Hydrogel Scaffold for Articular Cartilage Defect Repair. Adv Healthc Mater 2022; 11:e2101661. [PMID: 34969180 DOI: 10.1002/adhm.202101661] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/21/2021] [Indexed: 01/08/2023]
Abstract
After cartilage damage, a large number of monocytes/macrophages infiltrate into adjacent synovium and the resident macrophages in synovial tissue transform to activated macrophages (M1), which secrete pro-inflammatory cytokines to induce sustained inflammation and chondrocyte apoptotic. However, current clinical therapies for cartilage repair can rarely achieve long-term anti-inflammatory regulation and satisfactory outcomes. Herein, a platelet lysate-rich plasma macroporous hydrogel (PLPMH) scaffold with around 100 µm pore size and 1.25 MPa Young's modulus is developed to sustainedly recruit and polarize endogenous anti-inflammatory macrophages (M2) for improving cartilage defect repair. PLPMH scaffold can steadily release sphingosine1-phosphate and proteins via gradual degradation, thus inducing M2 macrophages migration or resting (M0) macrophages migration and then polarization to M2 phenotype, and improving the secretion of anti-inflammatory cytokines. Furthermore, PLPMH scaffold exhibits negligible inflammatory responses in vivo and promotes endogenous M2 macrophage infiltration in large numbers and long-time duration to provide a local anti-inflammatory microenvironment, which even lasts for 42 d. In a rabbit model of cartilage defect, PLPMH scaffold increases the ratio of M2 macrophages and improves cartilage tissue regeneration. These studies support that PLPMH scaffold may have a great potential in articular cartilage tissue engineering by providing an anti-inflammatory and pro-regenerative microenvironment.
Collapse
Affiliation(s)
- Xiaoyun Pan
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Key Laboratory of Orthopaedics of Zhejiang Province Wenzhou Medical University Wenzhou Zhejiang 325000 China
| | - Shanshan Yuan
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | - Xiaojie Xun
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | | | - Xinghe Xue
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Key Laboratory of Orthopaedics of Zhejiang Province Wenzhou Medical University Wenzhou Zhejiang 325000 China
| | - Changhuan Zhang
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | - Jilong Wang
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| | - Junjie Deng
- Department of Orthopaedics The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Key Laboratory of Orthopaedics of Zhejiang Province Wenzhou Medical University Wenzhou Zhejiang 325000 China
- Wenzhou Institute University of Chinese Academy Sciences Wenzhou Zhejiang 325000 China
| |
Collapse
|
42
|
Chen W, Zheng H, Zhang X, Xu Y, Fu Z, Ji X, Wei C, An G, Tan M, Zhou M. Columbianetin alleviates lipopolysaccharides (LPS)-induced inflammation and apoptosis in chondrocyte through activation of autophagy by inhibiting serum and glucocorticoid-induced protein kinase 1 (SGK1) expression. Bioengineered 2022; 13:4051-4062. [PMID: 35129051 PMCID: PMC8973585 DOI: 10.1080/21655979.2022.2032970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of articular cartilage involving the entire joint tissue. Columbianetin (CBT) is a major active compound of radix angelicae pubescentis, which is used in the treatment of OA. This paper attempts to explore the role of CBT in OA. Lipopolysaccharides (LPS) was used to induce mouse chondrocytes ATDC5. The effect of CBT on cell viability in ATDC5 cells with or without LPS induction was determined by CCK-8 and LDH kits. The inflammatory response was evaluated using ELISA kits. Apoptosis in LPS-induced ATDC5 cells were examined by TUNEL staining. The expression of apoptosis and autophagy-related proteins was tested with Western blot. The relationship between CBT and serum and glucocorticoid-induced protein kinase 1 (SGK1) was examined by RT-qPCR, Western blot, and molecular docking. After SGK1 overexpression or addition of the autophagy inhibitor 3-methyladenine (3 MA), the above experiments were done again. Results revealed that CBT increased LPS-induced decrease in ATDC5 cell viability. CBT inhibited inflammation triggered by LPS, evidenced by reduced levels of TNF-α, IL-6 and IL-1β. Cell apoptosis was attenuated following CBT adding in ATDC5 cells exposed to LPS, accompanied by upregulated Bcl-2 expression and downregulated Bax and cleaved caspase 3 expression. In addition, CBT elevated Beclin1 and LC3II/LC3I expression but decreased p62 expression. Additionally, CBT inhibited SGK1 expression. However, SGK1 overexpression or 3 MA reversed the effects of CBT on LPS-induced loss of ATDC5 cell viability, inflammation, apoptosis and autophagy. Collectively, CBT could improve OA through the activation of chondrocyte autophagy by suppressing SGK1 expression.
Collapse
Affiliation(s)
- Wei Chen
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Haotian Zheng
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xuan Zhang
- Department of Oncology, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Yude Xu
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Zhibin Fu
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Xing Ji
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Changhao Wei
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Guoyao An
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Mingyuan Tan
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| | - Mingwang Zhou
- Department of Orthopaedics, Traditional Chinese Medical Hospital of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
43
|
Tang L, Sim I, Moqbel S, Wu L. Dapansutrile ameliorated chondrocyte inflammation and osteoarthritis through suppression of MAPK signaling pathway. Hum Exp Toxicol 2022; 41:9603271221145401. [PMID: 36508695 DOI: 10.1177/09603271221145401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Osteoarthritis (OA) is one of the most common joint diseases in the elderly population. Proinflammatory cytokines, such as Interleukin-1β (IL-1β), play an important role in the development and progression of OA. Dapansutrile is a specific inhibitor of the NOD-like receptor protein 3 (NLRP3) inflammasome and exhibits anti-inflammatory properties. METHODS In this study, we investigated the protective effect and the underlying mechanism of dapansutrile on cartilage degeneration in vitro and in vivo. In the present study, chondrocytes were isolated from rats and then were treated with dapansutrile. After that, the expression of (Cox-2, inducible nitric oxide synthase (iNOS), Mmp-3, Mmp-9, Mmp-13 and IL-10) were evaluated at RNA level, then the expression of (COX-2, MMP-3, MMP-9, MMP-13, SOX-9 and COL2) were evaluated at protein level. Subsequently, the activation of the mitogen-activated protein kinase (MAPK) pathway was tested using western blotting (WB). Additionally, the rat OA model was developed to evaluate the protective effects of dapansutrile in vivo. RESULTS The results showed that dapansutrile had no obvious cytotoxicity on rat chondrocytes at 24 h (0, 1, 2, 5 and 10 μM). Dapansutrile significantly decreased IL-1β-induced upregulation of COX2, iNOS, matrix metalloproteinase 3 (MMP3), 9 (MMP9) and 13 (MMP13), and reversed IL-1β-induced the downregulation of IL-10, SOX9 and COL2. Dapansutrile also inhibited IL-1β-induced upregulation of the MAPK signaling pathway by downregulating the expression levels of phospho-ERK, and phospho-P38 in a concentration dependent manner. In addition, dapansutrile exhibited protective effects in rat OA model with lower Mankin's score and Osteoarthritis Research Society International (OARSI) score. CONCLUSION Our study suggested that dapansutrile effectively inhibited chondrocyte inflammation by suppressing MAPK signaling pathway in vitro, and ameliorated cartilage degeneration in vivo, indicating an anti-inflammatory effect in OA treatment.
Collapse
Affiliation(s)
- L Tang
- Department of Emergency Medicine, 89681The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou City, Zhejiang, China
| | - I Sim
- Department of Oncology, Clinical Institute, 89681Pyongyang Medical University, Pyongyang, Democratic People's Republic of Korea.,Department of Orthopedic Surgery, 89681The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, China
| | - Saa Moqbel
- Department of Emergency Medicine, 89681The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou City, Zhejiang, China
| | - L Wu
- Department of Orthopedic Surgery, 89681The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, China
| |
Collapse
|
44
|
Qin Y, Li J, Zhou Y, Yin C, Li Y, Chen M, Du Y, Li T, Yan J. Apolipoprotein D as a Potential Biomarker and Construction of a Transcriptional Regulatory-Immune Network Associated with Osteoarthritis by Weighted Gene Coexpression Network Analysis. Cartilage 2021; 13:1702S-1717S. [PMID: 34719950 PMCID: PMC8808834 DOI: 10.1177/19476035211053824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE Synovial inflammation influences the progression of osteoarthritis (OA). Herein, we aimed to identify potential biomarkers and analyze transcriptional regulatory-immune mechanism of synovitis in OA using weighted gene coexpression network analysis (WGCNA). DESIGN A data set of OA synovium samples (GSE55235) was analyzed based on WGCNA. The most significant module with OA was identified and function annotation of the module was performed, following which the hub genes of the module were identified using Pearson correlation and a protein-protein interaction network was constructed. A transcriptional regulatory network of hub genes was constructed using the TRRUST database. The immune cell infiltration of OA samples was evaluated using the single-sample Gene Set Enrichment Analysis (ssGSEA) method. The hub genes coexpressed in multiple tissues were then screened out using data sets of synovium, cartilage, chondrocyte, subchondral bone, and synovial fluid samples. Finally, transcriptional factors and coexpressed hub genes were validated via experiments. RESULTS The turquoise module of GSE55235 was identified via WGCNA. Functional annotation analysis showed that "mineral absorption" and "FoxO signaling pathway" were mostly enriched in the module. JUN, EGR1, FOSB, and KLF4 acted as central nodes in protein-protein interaction network and transcription factors to connect several target genes. "Activated B cell," "activated CD4T cell," "eosinophil," "neutrophil," and "type 17 T helper cell" showed high immune infiltration, while FOSB, KLF6, and MYBL2 showed significant negative correlation with type 17 T helper cell. CONCLUSIONS Our results suggest that the expression level of apolipoprotein D (APOD) was correlated with OA. Furthermore, transcriptional regulatory-immune network was constructed, which may contribute to OA therapy.
Collapse
Affiliation(s)
- Yong Qin
- Department of Orthopedics Surgery, The
Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia Li
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yonggang Zhou
- Department of Orthopedics Surgery, The
Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chengliang Yin
- Medical Big Data Research Center,
Medical Innovation Research Division of Chinese PLA General Hospital, Beijing,
China,National Engineering Laboratory for
Medical Big Data Application Technology, Chinese PLA General Hospital, Beijing,
China,Faculty of Medicine, Macau University
of Science and Technology, Macau, China
| | - Yi Li
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ming Chen
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yinqiao Du
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Tiejian Li
- Department of Orthopedics Surgery, The
First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jinglong Yan
- Department of Orthopedics Surgery, The
Second Affiliated Hospital of Harbin Medical University, Harbin, China,Jinglong Yan, Department of Orthopedics
Surgery, The Second Affiliated Hospital of Harbin Medical University, No.246
Xuefu Road, Harbin 150086, China.
| |
Collapse
|
45
|
Cai C, Zhang Y, Peng X. Knocking down Sterol regulatory element binding protein 2 (SREBF2) inhibits the Serine Protease 8 (PRSS8) /sodium channel epithelial 1alpha subunit (SCNN1A) axis to reduce the cell proliferation, migration and epithelial-mesenchymal transformation of ovarian cancer. Bioengineered 2021; 12:9390-9400. [PMID: 34823420 PMCID: PMC8809903 DOI: 10.1080/21655979.2021.1978615] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The pathogenesis of ovarian cancer (OC) is complex. Serine Protease 8 (PRSS8) is a potential biomarker for early detection of OC. Multiple databases were used to predict the expression of PRSS8, Sterol regulatory element binding protein (SREBP) and sodium channel epithelial 1alpha subunit (SCNN1A) in OC patients and to detect the relationship among the three. The expressions of PRSS8, SREBF2, SCNN1A and related factors of the pathway were detected by RT-qPCR and Western blot. The cell transfection was used to overexpress or inhibit the expression of PRSS8 and SREBF2, so as to explore its mechanism. MTT assay and Colony formation assay were used to detect cell proliferation. The Transwell and Wound Healing assays were utilized to measure cell invasion and migration. We have further confirmed cell-level studies in animals. We found that PRSS8 expression was up-regulated in OC patients and cell lines. Knocking down PRSS8 reduced the proliferation, migration and epithelial-mesenchymal transition (EMT) of OC cells, which was realized by SREBF2 transcriptional regulation. Knocking down SREBF2 reduced PRSS8 and then inhibited the expression of SCNN1A, thus affecting the proliferation, migration and EMT of OC cells. These results also applied to animals experiments. In conclusion, SREBF2 activates the PRSS8/SCNN1A axis to accelerate cell proliferation, migration and EMT of OC.
Collapse
Affiliation(s)
- Chunyan Cai
- Department of Gynaecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yumei Zhang
- Department of Gynaecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Xing Peng
- Department of Gynaecology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
46
|
Lara-Barba E, Araya MJ, Hill CN, Bustamante-Barrientos FA, Ortloff A, García C, Galvez-Jiron F, Pradenas C, Luque-Campos N, Maita G, Elizondo-Vega R, Djouad F, Vega-Letter AM, Luz-Crawford P. Role of microRNA Shuttled in Small Extracellular Vesicles Derived From Mesenchymal Stem/Stromal Cells for Osteoarticular Disease Treatment. Front Immunol 2021; 12:768771. [PMID: 34790203 PMCID: PMC8591173 DOI: 10.3389/fimmu.2021.768771] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/14/2021] [Indexed: 12/18/2022] Open
Abstract
Osteoarticular diseases (OD), such as rheumatoid arthritis (RA) and osteoarthritis (OA) are chronic autoimmune/inflammatory and age-related diseases that affect the joints and other organs for which the current therapies are not effective. Cell therapy using mesenchymal stem/stromal cells (MSCs) is an alternative treatment due to their immunomodulatory and tissue differentiation capacity. Several experimental studies in numerous diseases have demonstrated the MSCs’ therapeutic effects. However, MSCs have shown heterogeneity, instability of stemness and differentiation capacities, limited homing ability, and various adverse responses such as abnormal differentiation and tumor formation. Recently, acellular therapy based on MSC secreted factors has raised the attention of several studies. It has been shown that molecules embedded in extracellular vesicles (EVs) derived from MSCs, particularly those from the small fraction enriched in exosomes (sEVs), effectively mimic their impact in target cells. The biological effects of sEVs critically depend on their cargo, where sEVs-embedded microRNAs (miRNAs) are particularly relevant due to their crucial role in gene expression regulation. Therefore, in this review, we will focus on the effect of sEVs derived from MSCs and their miRNA cargo on target cells associated with the pathology of RA and OA and their potential therapeutic impact.
Collapse
Affiliation(s)
- Eliana Lara-Barba
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Charlotte Nicole Hill
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.,Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Ciencias Biológicas, Millennium Institute for Immunology and Immunotherapy, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Felipe Galvez-Jiron
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Gabriela Maita
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.,Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Farida Djouad
- Institute for Regenerative Medicine and Biotherapy (IRMB), Univ Montpellier, Institut national de la santé et de la recherche médicale (INSERM), Montpellier, France
| | - Ana María Vega-Letter
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de Los Andes, Santiago, Chile.,IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| |
Collapse
|
47
|
Lin YY, Ko CY, Liu SC, Wang YH, Hsu CJ, Tsai CH, Wu TJ, Tang CH. miR-144-3p ameliorates the progression of osteoarthritis by targeting IL-1β: Potential therapeutic implications. J Cell Physiol 2021; 236:6988-7000. [PMID: 33772768 DOI: 10.1002/jcp.30361] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 12/16/2022]
Abstract
The pro-inflammatory cytokine interleukin 1 beta (IL-1β) plays a critical role in osteoarthritis (OA) disease pathogenesis. MicroRNA (miRNA) activity is related to inflammation in OA and some miRNAs specifically regulate IL-mediated degradation of cartilage type II collagen. Previous studies have indicated that miR-144-3p is a useful target in the regulation of pro-inflammatory cytokines in different diseases. However, the role of miR-144-3p in OA is unclear. In this study, we observed a negative correlation between miR-144-3p and IL-1β expression in OA. miR-144-3p mimic transfection of OA synovial fibroblasts downregulated levels of IL-1β expression, while blocking the MAPK, PI3K/Akt, and NF-κB signaling pathways relating to IL-1β production, and effectively increased miR-144-3p expression in OASFs. Findings from an anterior cruciate ligament transection rat model revealed that administration of miR-144-3p mimic effectively ameliorated OA progression and reduced the numbers of IL-1β-positive cells in synovial tissue. This study suggests that miR-144-3p is a useful therapeutic target in OA disease.
Collapse
Affiliation(s)
- Yen-You Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Yu-Han Wang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chin-Jung Hsu
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Sports Medicine, China Medical University, Taichung, Taiwan
| | - Tsung-Ju Wu
- Department of Physical Medicine and Rehabilitation, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
48
|
Li Z, Wang J, Ma Y. Montelukast attenuates interleukin IL-1β-induced oxidative stress and apoptosis in chondrocytes by inhibiting CYSLTR1 (Cysteinyl Leukotriene Receptor 1) and activating KLF2 (Kruppel Like Factor 2). Bioengineered 2021; 12:8476-8484. [PMID: 34565285 PMCID: PMC8806840 DOI: 10.1080/21655979.2021.1984003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Montelukast is a cysteinyl leukotriene receptor 1 (CysLTR1) antagonist widely used to suppress the inflammatory response in asthma and allergic rhinitis. This study aimed to investigate the potential impacts of montelukast on osteoarthritis (OA) progression. To determine the role of montelukast in OA, the expression of CysLTR1 was first examined by quantitative reverse transcription PCR (RT-qPCR) and western blot in IL-1β-induced ATDC5 cells treated with or without montelukast. Subsequently, the impacts of montelukast on cell viability and oxidative stress were measured by Cell-Counting-Kit-8 (CCK-8), commercial kits and western blot. Oxidative stress-related protein expressions were determined by western blot analysis in Il-1β-induced ATDC5 cells. Cell apoptosis and cartilage degradation were examined by TdT-mediated dUTP Nick-End Labeling (TUNEL) assay, western blot and RT-qPCR. KLF2 expression was measured in IL-1β-induced ATDC5 cells treated with montelukast. After interference with small interfering RNA (siRNA)-KLF2 in ATDC5 cells, the loss-of-function assays were also performed in same ways. CysLTR1 expression was elevated in IL-1β-induced ATDC5 cells but inhibited significantly by montelukast. Montelukast attenuated the oxidative stress and apoptosis, improved cell viability. Moreover, montelukast enhanced KLF2 expression. After transfected with siRNA-KLF2, montelukast attenuated cell injury, oxidative stress, apoptosis and cartilage degradation in IL-1β-induced ATDC5 cells by activating KLF2.In summary, this work elaborates the evidence that montelukast could attenuate oxidative stress and apoptosis in IL-1β-induced chondrocytes by inhibiting CysLTR1 and activating KLF2, which can guide the therapeutic strategies of montelukast for OA development in the future.
Collapse
Affiliation(s)
- Zongwei Li
- School of Pharmaceutical Engineering, Guangdong Food and Drug Vocational College, Guangzhou City, Guangdong Province, China
| | - Jianming Wang
- School of Pharmaceutical Engineering, Guangdong Food and Drug Vocational College, Guangzhou City, Guangdong Province, China
| | - Yumin Ma
- Department of Pharmaceutical Machinery, Maternal and Child Health and Family Planning Technical Service Center, Wuwei City, Gansu Province, China
| |
Collapse
|
49
|
Mei X, Din H, Zhao J, Tong J, Zhu W. Transcription factor Krüppel-like factor 5-regulated N-myc downstream-regulated gene 2 reduces IL-1β-induced chondrocyte inflammatory injury and extracellular matrix degradation. Bioengineered 2021; 12:7020-7032. [PMID: 34551684 PMCID: PMC8806548 DOI: 10.1080/21655979.2021.1971483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Previous research has identified N-myc downstream-regulated gene 2 (NDRG2) as one of the differentially expressed genes common to rat models of osteoarthritis (OA) and human OA. The purpose of this study was to investigate the role of NDRG2 in OA. In this study, an in vitro OA model was constructed by challenging ATDC5 chondrocytes with 10 ng/ml IL-1β. After transfection of pcDNA3.1(+)/NDRG2, qPCR and western blot were performed to assay NDRG2 expression. The analyses of cell viability, apoptosis and inflammatory molecule expression were employed respectively by CCK-8, TUNEL and ELISA. The protein expression related to apoptosis, inflammation or extracellular matrix (ECM) degradation was detected by western blot. The binding of Krüppel-like factor 5 (KLF5) to NDRG2 promoter was verified by means of dual-luciferase reporter assay. After overexpression of both NDRG2 and KLF5 in IL-1β-stimulated ATDC5 chondrocytes, corresponding assays were performed to examine cell viability, apoptosis, inflammatory response and ECM degradation. In ATDC5 chondrocytes challenged with IL-1β, NDRG2 expression was much lower than that in the control group, whereas it’s overexpression helped restored cell viability and reduce cell apoptosis, inflammatory response and ECM degradation. It was also observed that KLF5 expression was decreased in IL-1β-stimulated ATDC5 chondrocytes, and that KLF5 bound to the NDRG2 promoter. Importantly, overexpressing KLF5 could reverse the protective effect of NDRG2 overexpression on IL-1β-stimulated ATDC5. Overall, NDRG2 could be transcriptionally regulated by transcription factor KLF5 and may play a protective role against chondrocyte the inflammatory response and ECM degradation in OA.
Collapse
Affiliation(s)
- Xiaoliang Mei
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, People's Republic of China
| | - Hao Din
- Department of Orthopedics, Jinling Hospital, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jianning Zhao
- Department of Orthopedics, Jinling Hospital, Jinling Clinical Medical College of Nanjing Medical University, Nanjing, People's Republic of China
| | - Jian Tong
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, People's Republic of China
| | - Wei Zhu
- Department of Orthopedics, Taizhou Clinical Medical School of Nanjing Medical University (Taizhou People's Hospital), Taizhou, People's Republic of China
| |
Collapse
|
50
|
Lee KT, Chen BC, Liu SC, Lin YY, Tsai CH, Ko CY, Tang CH, Tung KC. Nesfatin-1 facilitates IL-1β production in osteoarthritis synovial fibroblasts by suppressing miR-204-5p synthesis through the AP-1 and NF-κB pathways. Aging (Albany NY) 2021; 13:22490-22501. [PMID: 34560673 PMCID: PMC8507299 DOI: 10.18632/aging.203559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/07/2021] [Indexed: 12/11/2022]
Abstract
The progression of osteoarthritis (OA) is mediated by adipokines, one of which is nesfatin-1, which is responsible for the production of inflammatory cytokines. However, how this molecule may affect the synthesis of the proinflammatory cytokine interleukin 1 beta (IL-1β) in OA is unclear. Our analyses of records from the Gene Expression Omnibus (GEO) dataset and clinical specimens of synovial tissue revealed higher levels of nesfatin-1 and IL-1β in OA samples compared with normal healthy tissue. We found that nesfatin-1 facilitates IL-1β synthesis in human OA synovial fibroblasts (OASFs) and suppresses the generation of micro-RNA (miR)-204-5p, as the miR-204-5p levels in OA patients were lower than those in healthy controls. Nesfatin-1-induced stimulation of IL-1β in human OASFs occurred via the suppression of miR-204-5p synthesis by the PI3K, Akt, AP-1 and NF-κB pathways. We suggest that nesfatin-1 is worth targeting in OA treatment.
Collapse
Affiliation(s)
- Kun-Tsan Lee
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan.,Department of Orthopedics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Bo-Cheng Chen
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Yen-You Lin
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Kwong-Chung Tung
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|