1
|
Peng M, Shen G, Tu Q, Zhang W, Wang J. Nuciferine ameliorates osteoarthritis: An in vitro and in vivo study. Int Immunopharmacol 2024; 142:113098. [PMID: 39321708 DOI: 10.1016/j.intimp.2024.113098] [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: 03/17/2024] [Revised: 07/19/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
Osteoarthritis (OA) is the most common musculoskeletal disease and a leading cause of pain and disability. A key hallmark of OA is cartilage degradation, which occurs due to an imbalance between the synthesis and degradation of the extracellular matrix (ECM). Interleukin-1β(IL-1β) has been reported to regulate ECM metabolism. Nuciferine (Nuc), a natural peptide extracted from the lotus leaf, possesses several significant pharmacological properties. However, the anti-inflammation of Nuc in OA has not been reported. In this study, ELISA and Western blot analyses were used to measure the production of inflammatory mediators in IL-1β-Induced mouse chondrocytes. Additionally, mice with or without surgical destabilization of the medial meniscus (DMM) were treated with intra-articular injection of Nuc. We found that Nuc significantly reduces the level of iNOS, PEG2, and IL-6 in IL-1β-induced chondrocytes. Furthermore, Nuc can ameliorate the development of OA in mice. Mechanistically, we found that the chondrocyte-protective effects of Nuc occur via the PTEN/NF-κB pathway. These findings suggest that Nuc could be a potential therapeutic agent for improving OA development.
Collapse
Affiliation(s)
- Maoxiu Peng
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Guangjie Shen
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Qiming Tu
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Weihao Zhang
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Juncheng Wang
- Department of Orthopaedic Surgery, The Third Hospital Affiliated to Wenzhou Medical University, Rui'an 325200, China; Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.
| |
Collapse
|
2
|
Wang D, Cao L, Zhang H, Wang X, You W. LncRNA NKILA attenuates the progression of osteoarthritis through the targeted inhibition of the NF-κB pathway. Int Immunopharmacol 2024; 143:113417. [PMID: 39447414 DOI: 10.1016/j.intimp.2024.113417] [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: 08/14/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Interleukin-1β (IL-1β) plays a crucial role in cartilage degeneration by inducing inflammatory cascades in chondrocytes, impairing their normal biological functions. Long non-coding RNA NKILA (lncRNA NKILA) has been implicated in osteoarthritis (OA), but its specific molecular mechanisms remain unclear. This study aims to elucidate the function and molecular regulatory mechanism of lncRNA NKILA in articular chondrocytes under IL-1β stimulation. METHODS Primary human articular chondrocytes were cultured to investigate the effects of IL-1β on chondrocyte proliferation, apoptosis, and extracellular matrix metabolism. We employed Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR), western blot, flow cytometry, immunofluorescence, and nuclear mass separation assays to explore the interaction between lncRNA NKILA and the NFκB signaling pathway. Additionally, animal experiments were conducted to evaluate the therapeutic potential of modulating lncRNA NKILA expression in vivo. RESULTS IL-1β treatment led to decreased chondrocyte proliferation and increased apoptosis. Our study demonstrated that IL-1β downregulates lncRNA NKILA, which weakens its inhibitory effect on the NFκB (Nuclear Factor Kappa B) signaling pathway. This downregulation results in increased NFκB activity and exacerbates chondrocyte degeneration. Notably, the upregulation of lncRNA NKILA significantly alleviated OA symptoms, indicating that NKILA could be a promising therapeutic target. CONCLUSION IL-1β reduces lncRNA NKILA expression, weakening its inhibition of NFκB signaling and promoting articular chondrocyte degeneration. Enhancing lncRNA NKILA expression offers a promising approach to mitigating OA, suggesting that NKILA could serve as a potential therapeutic target for OA treatment.
Collapse
Affiliation(s)
- Dongmei Wang
- Department of Pharmacy, The Second Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Lixin Cao
- Department of Orthopaedics, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, China
| | - Honglian Zhang
- College of Pharmacy, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Xuefeng Wang
- Department of Orthopaedics, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, China
| | - Weifu You
- Department of Orthopaedics, The First Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161000, China.
| |
Collapse
|
3
|
Singer J, Knezic N, Layne J, Gohring G, Christiansen J, Rothrauff B, Huard J. Enhancing Cartilage Repair: Surgical Approaches, Orthobiologics, and the Promise of Exosomes. Life (Basel) 2024; 14:1149. [PMID: 39337932 PMCID: PMC11432843 DOI: 10.3390/life14091149] [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: 07/28/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Treating cartilage damage is challenging as its ability for self-regeneration is limited. Left untreated, it can progress to osteoarthritis (OA), a joint disorder characterized by the deterioration of articular cartilage and other joint tissues. Surgical options, such as microfracture and cell/tissue transplantation, have shown promise as techniques to harness the body's endogenous regenerative capabilities to promote cartilage repair. Nonetheless, these techniques have been scrutinized due to reported inconsistencies in long-term outcomes and the tendency for the defects to regenerate as fibrocartilage instead of the smooth hyaline cartilage native to joint surfaces. Orthobiologics are medical therapies that utilize biologically derived substances to augment musculoskeletal healing. These treatments are rising in popularity because of their potential to enhance surgical standards of care. More recent developments in orthobiologics have focused on the role of exosomes in articular cartilage repair. Exosomes are nano-sized extracellular vesicles containing cargo such as proteins, lipids, and nucleic acids, and are known to facilitate intercellular communication, though their regenerative potential still needs to be fully understood. This review aims to demonstrate the advancements in cartilage regeneration, highlight surgical and biological treatment options, and discuss the recent strides in understanding the precise mechanisms of action involved.
Collapse
Affiliation(s)
- Jacob Singer
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Noah Knezic
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jonathan Layne
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Greta Gohring
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Jeff Christiansen
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ben Rothrauff
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
4
|
Chen R, Tong Y, Hu X, Wang W, Liao F. circSLTM knockdown attenuates chondrocyte inflammation, apoptosis and ECM degradation in osteoarthritis by regulating the miR-515-5p/VAPB axis. Int Immunopharmacol 2024; 138:112435. [PMID: 38981227 DOI: 10.1016/j.intimp.2024.112435] [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: 02/24/2024] [Revised: 05/20/2024] [Accepted: 06/05/2024] [Indexed: 07/11/2024]
Abstract
Osteoarthritis (OA) is a prevalent joint disorder characterized by cartilage degeneration. Circular RNAs (circRNAs) have emerged as pivotal players in OA progression, orchestrating various biological processes such as proliferation, apoptosis, inflammation, and extracellular matrix (ECM) reorganization. Among these circRNAs, circSLTM exhibits aberrant expression in OA, yet its precise regulatory mechanism remains elusive. This study aimed to elucidate the regulatory mechanisms of circSLTM in OA pathogenesis, with a focus on its role as a competing endogenous RNA (ceRNA). Human cartilage tissues were procured from both OA patients and non-OA individuals, while human chondrocyte cells were subjected to lipopolysaccharide (LPS) treatment to mimic OA-like conditions. Our findings revealed upregulation of circSLTM in OA patients and LPS-treated chondrocytes. Loss-of-function assays were conducted, demonstrating that silencing circSLTM via shRNAs mitigated LPS-induced effects on chondrocytes, as evidenced by enhanced proliferation, reduced apoptosis, and inflammatory factors, and altered expression of extracellular matrix proteins. Further exploration into the regulatory mechanism of circSLTM unveiled its interaction with microRNA-515-5p (miR-515-5p) to modulate vesicle-associated membrane protein (VAPB) expression in chondrocytes. VAPB, also upregulated in OA, was positively regulated by circSLTM. Rescue assays corroborated that VAPB overexpression reinstated the protective effects of circSLTM knockdown on LPS-treated chondrocytes. Moreover, concurrent knockdown of both circSLTM and VAPB demonstrated synergistic protection against LPS-induced chondrocyte injury. Additionally, we delineated that LPS triggered the activation of the NF-κB pathway in chondrocytes, which was counteracted by circSLTM silencing. To assess the effects of circSLTM on OA in vivo, anterior cruciate ligament transection (ACLT) mouse models were established, revealing that circSLTM deficiency ameliorated cartilage defects in vivo. In conclusion, circSLTM exacerbates osteoarthritis progression by orchestrating the miR-515-5p/VAPB axis and activating the NF-κB pathway, providing novel insights for targeted therapy in OA management.
Collapse
Affiliation(s)
- Rijiang Chen
- Department of Orthopedics, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian 364000, China.
| | - Yan Tong
- Department of Endocrine, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian 364000, China.
| | - Xiunian Hu
- Department of Orthopedics, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian 364000, China.
| | - Wantao Wang
- Department of Orthopedics, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian 364000, China.
| | - Fake Liao
- Department of Orthopedics, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian 364000, China.
| |
Collapse
|
5
|
Xu B, Xu Y, Kong J, Liu Y, Zhang L, Shen F, Wang J, Shen X, Chen H. Chrysin mitigated neuropathic pain and peripheral sensitization in knee osteoarthritis rats by repressing the RAGE/PI3K/AKT pathway regulated by HMGB1. Cytokine 2024; 180:156635. [PMID: 38749277 DOI: 10.1016/j.cyto.2024.156635] [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: 02/05/2024] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a chronic progressive osteoarthropathy. Chrysin's anti-KOA action has been demonstrated, however more research is needed to understand how chrysin contributes to KOA. METHODS LPS/ATP-induced macrophages transfected with or without HMGB1 overexpression underwent 5 μg/mL chrysin. The cell viability and macrophage pyroptosis were examined by cell counting kit-8 and flow cytometer. In vivo experiments, rats were injected with 1 mg monosodium iodoacetate by the infrapatellar ligament of the bilateral knee joint to induce KOA. The histological damage was analyzed by Safranin O/Fast Green staining and hematoxylin and eosin staining. The PWT, PWL and inflammatory factors were analyzed via Von-Frey filaments, thermal radiometer and ELISA. Immunofluorescence assay examined the expressions of CGRP and iNOS. The levels of HMGB1/RAGE-, NLRP3-, PI3K/AKT- and neuronal ion channel-related markers were examined by qPCR and western blot. RESULTS Chrysin alleviated macrophage pyroptosis by inhibiting HMGB1 and the repression of chrysin on HMGB1/RAGE pathway and ion channel activation was reversed by overexpressed HMGB1. HMGB1 facilitated neuronal ion channel activation through the RAGE/PI3K/AKT pathway. Chrysin could improve the pathological injury of knee joints in KOA rats. Chrysin suppressed the HMGB1-regulated RAGE/PI3K/AKT pathway, hence reducing KOA damage and peripheral sensitization. CONCLUSION Chrysin mitigated neuropathic pain and peripheral sensitization in KOA rats by repressing the RAGE/PI3K/AKT pathway modulated by HMGB1.
Collapse
Affiliation(s)
- Bo Xu
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China
| | - Yue Xu
- Department of Orthopedics and Traumatology, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu 215500, Jiangsu Province, PR China
| | - Jian Kong
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China
| | - Yujiang Liu
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China
| | - Long Zhang
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China
| | - Fan Shen
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China
| | - Jiangping Wang
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China
| | - Xiaofeng Shen
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China.
| | - Hua Chen
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, Jiangsu Province, PR China.
| |
Collapse
|
6
|
Huang F, Su Z, Yang J, Zhao X, Xu Y. Knocking-down long non-coding RNA LINC01094 prohibits chondrocyte apoptosis via regulating microRNA-577/metal-regulatory transcription factor 1 axis. J Orthop Surg (Hong Kong) 2024; 32:10225536241254588. [PMID: 38758016 DOI: 10.1177/10225536241254588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2024] Open
Abstract
PURPOSE The abnormal function and survival of chondrocytes result in articular cartilage failure, which may accelerate the onset and development of osteoarthritis (OA). This study is aimed to investigate the role of LINC01094 in chondrocyte apoptosis. METHODS The viability and apoptosis of lipopolysaccharide (LPS)-induced chondrocytes were evaluated through CCK-8 assay and flow cytometry analysis, respectively. The expression levels of LINC01094, miR-577 and MTF1 were detected by qRT-PCR. Dual luciferase reporter tests were implemented for the verification of targeted relationships among them. Western blotting was employed to measure the levels of pro-apoptotic proteins (Caspase3 and Caspase9). RESULTS The viability of LPS-induced chondrocytes was overtly promoted by loss of LINC01094 or miR-577 upregulation, but could be repressed via MTF1 overexpression. The opposite results were observed in apoptosis rate and the levels of Caspase3 and Caspase9. LINC01094 directly bound to miR-577, while MTF1 was verified to be modulated by miR-577. Both LINC01094 and MTF1 were at high levels, whereas miR-577 was at low level in OA synovial fluid and LPS-induced chondrocytes. Furthermore, the highly expressed miR-577 abolished the influences of MTF1 overexpression on LPS-induced chondrocytes. CONCLUSIONS Silencing of LINC01094 represses the apoptosis of chondrocytes through upregulating miR-577 expression and downregulating MTF1 levels, providing a preliminary insight for the treatment of OA in the future.
Collapse
Affiliation(s)
- Feiri Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, The Third Affiliated Hospital of Shanghai University; The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Zhongliang Su
- Department of Orthopedics, The Third Affiliated Hospital of Shanghai University; The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Jie Yang
- Department of Orthopedics, The Third Affiliated Hospital of Shanghai University; The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Xizhen Zhao
- Department of Orthopedics, The Third Affiliated Hospital of Shanghai University; The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Yaozeng Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Wu J, Huang H, Gong L, Tian X, Peng Z, Zhu Y, Wang W. A Flavonoid Glycoside Compound from Siraitia grosvenorii with Anti-Inflammatory and Hepatoprotective Effects In Vitro. Biomolecules 2024; 14:450. [PMID: 38672467 PMCID: PMC11048398 DOI: 10.3390/biom14040450] [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: 03/02/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammation is a pivotal factor in the development and advancement of conditions like NAFLD and asthma. Diet can affect several phases of inflammation and significantly influence multiple inflammatory disorders. Siraitia grosvenorii, a traditional Chinese edible and medicinal plant, is considered beneficial to health. Flavonoids can suppress inflammatory cytokines, which play a crucial role in regulating inflammation. In the present experiments, kaempferol 3-O-α-L-rhamnoside-7-O-β-D-xylosyl(1→2)-O-α-L-rhamnoside (SGPF) is a flavonoid glycoside that was first isolated from S. grosvenorii. A series of experimental investigations were carried out to investigate whether the flavonoid component has anti-inflammatory and hepatoprotective effects in this plant. The researchers showed that SGPF has a stronger modulation of protein expression in LPS-induced macrophages (MH-S) and OA-induced HepG2 cells. The drug was dose-dependent on cells, and in the TLR4/NF-κB/MyD88 pathway and Nrf2/HO-1 pathway, SGPF regulated all protein expression. SGPF has a clear anti-inflammatory and hepatoprotective function in inflammatory conditions.
Collapse
Affiliation(s)
- Juanjiang Wu
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huaxue Huang
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Hunan Huacheng Biotech, Inc., High-Tech Zone, Changsha 410205, China;
| | - Limin Gong
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xing Tian
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
| | - Zhi Peng
- Hunan Huacheng Biotech, Inc., High-Tech Zone, Changsha 410205, China;
| | - Yizhun Zhu
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
| | - Wei Wang
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
8
|
Papathanasiou I, Balis C, Destounis D, Mourmoura E, Tsezou A. NEAT1-mediated miR-150-5p downregulation regulates b-catenin expression in OA chondrocytes. Funct Integr Genomics 2023; 23:246. [PMID: 37468759 DOI: 10.1007/s10142-023-01139-4] [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: 02/10/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023]
Abstract
We investigated the role of miR-150-5p in osteoarthritic (OA) chondrocytes, as well as the possible regulatory role of long non-coding RNAs (lncRNAs) in miR-150-5p expression. TargetScan, StarBase, DIANA-LncBase, and Open Targets databases were used to predict miR-150-5p target genes, lncRNAs/miR-150-5p interactions, and OA-related genes. Protein-protein interaction (PPI) network was constructed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). Gene ontology (GO) and pathway analysis were performed using Enrichr database. A publicly available RNA-seq dataset was retrieved to identify differentially expressed lncRNAs in damaged vs intact cartilage. We re-analyzed the retrieved RNA-seq data and revealed 177 differentially expressed lncRNAs in damage vs intact cartilage, including Nuclear Paraspeckle Assembly Transcript 1(NEAT1). MiR-150-5p, NEAT1, b-catenin, matrix metallopeptidase 13 (MMP-13), and ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS-5) expressions were assessed by reverse transcription-quantitative PCR (RT-qPCR) and western blot assay. Knockout and transfection experiments were conducted to investigate the role of NEAT1/miR-150-5p/b-catenin in cartilage degradation. Bioinformatics analysis revealed that b-catenin was an OA-related miR-150-5p target. MiR-150-5p overexpression in OA chondrocytes resulted in decreased expression of b-catenin, as well as MMP-13 and ADAMTS-5, both being Wnt/b-catenin downstream target genes. NEAT1/miR-150-5p interaction was predicted by bioinformatics analysis, while NEAT1 knockout led to increased expression of miR-150-5p in OA chondrocytes. Moreover, inhibition of miR-150-5p reversed the repressive effects of NEAT1 silencing in b-catenin expression in OA chondrocytes. Our results support a possible catabolic role of NEAT1/miR-150-5p interaction in OA progression by regulating b-catenin expression.
Collapse
Affiliation(s)
- Ioanna Papathanasiou
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Charalampos Balis
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Dimitrios Destounis
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Evanthia Mourmoura
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece
| | - Aspasia Tsezou
- Faculty of Medicine, Laboratory of Cytogenetics and Molecular Genetics, University of Thessaly, Biopolis, 41500, Larissa, Greece.
- Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece.
| |
Collapse
|
9
|
Noriega-González D, Caballero-García A, Roche E, Álvarez-Mon M, Córdova A. Inflammatory Process on Knee Osteoarthritis in Cyclists. J Clin Med 2023; 12:jcm12113703. [PMID: 37297897 DOI: 10.3390/jcm12113703] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/17/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Osteoarthritis is a disorder affecting the joints and is characterized by cellular stress and degradation of the extracellular matrix cartilage. It begins with the presence of micro- and macro-lesions that fail to repair properly, which can be initiated by multiple factors: genetic, developmental, metabolic, and traumatic. In the case of the knee, osteoarthritis affects the tissues of the diarthrodial joint, manifested by morphological, biochemical, and biomechanical modifications of the cells and the extracellular matrix. All this leads to remodeling, fissuring, ulceration, and loss of articular cartilage, as well as sclerosis of the subchondral bone with the production of osteophytes and subchondral cysts. The symptomatology appears at different time points and is accompanied by pain, deformation, disability, and varying degrees of local inflammation. Repetitive concentric movements, such as while cycling, can produce the microtrauma that leads to osteoarthritis. Aggravation of the gradual lesion in the cartilage matrix can evolve to an irreversible injury. The objective of the present review is to explain the evolution of knee osteoarthritis in cyclists, to show the scarce research performed in this particular field and extract recommendations to propose future therapeutic strategies.
Collapse
Affiliation(s)
- David Noriega-González
- Department of Surgery, Ophthalmology, Otorhinolaryngology and Physiotherapy, Faculty of Medicine, HVUV, 47003 Valladolid, Spain
| | - Alberto Caballero-García
- Department of Anatomy and Radiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, University of Valladolid, Campus Los Pajaritos, 42004 Soria, Spain
| | - Enrique Roche
- Department of Applied Biology-Nutrition and Institute of Bioengineering, Miguel Hernández University (UMH), 03202 Elche, Spain
- Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Internal Medicine, University of Alcalá de Henares, 28801 Alcalá de Henares, Spain
| | - Alfredo Córdova
- Department of Biochemistry, Molecular Biology and Physiology, Faculty of Health Sciences, GIR Physical Exercise and Aging, University of Valladolid, Campus Duques de Soria, 42004 Soria, Spain
| |
Collapse
|
10
|
Liu H, Guan H, He F, Song Y, Li F, Sun-Waterhouse D, Li D. Therapeutic actions of tea phenolic compounds against oxidative stress and inflammation as central mediators in the development and progression of health problems: A review focusing on microRNA regulation. Crit Rev Food Sci Nutr 2023; 64:8414-8444. [PMID: 37074177 DOI: 10.1080/10408398.2023.2202762] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Many health problems including chronic diseases are closely associated with oxidative stress and inflammation. Tea has abundant phenolic compounds with various health benefits including antioxidant and anti-inflammatory properties. This review focuses on the present understanding of the impact of tea phenolic compounds on the expression of miRNAs, and elucidates the biochemical and molecular mechanisms underlying the transcriptional and post-transcriptional protective actions of tea phenolic compounds against oxidative stress- and/or inflammation-mediated diseases. Clinical studies showed that drinking tea or taking catechin supplement on a daily basis promoted the endogenous antioxidant defense system of the body while inhibiting inflammatory factors. The regulation of chronic diseases based on epigenetic mechanisms, and the epigenetic-based therapies involving different tea phenolic compounds, have been insufficiently studied. The molecular mechanisms and application strategies of miR-27 and miR-34 involved in oxidative stress response and miR-126 and miR-146 involved in inflammation process were preliminarily investigated. Some emerging evidence suggests that tea phenolic compounds may promote epigenetic changes, involving non-coding RNA regulation, DNA methylation, histone modification, ubiquitin and SUMO modifications. However, epigenetic mechanisms and epigenetic-based disease therapies involving phenolic compounds from different teas, and the potential cross-talks among the epigenetic events, remain understudied.
Collapse
Affiliation(s)
- Hui Liu
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, P.R. China
| | - Hui Guan
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, P.R. China
| | - Fatao He
- All-China Federation of Supply & Marketing Co-operatives, Jinan Fruit Research Institute, Jinan, P.R. China
| | - Ye Song
- All-China Federation of Supply & Marketing Co-operatives, Jinan Fruit Research Institute, Jinan, P.R. China
| | - Feng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, P.R. China
| | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, P.R. China
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Dapeng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Nutrition and Human Health in Universities of Shandong, Taian, P.R. China
| |
Collapse
|
11
|
Ding Q, Zhang R, Sheng G, Wang T, Jing S, Ma T, Wang S, Zhao H, Wu H, Li W. Dioscin alleviates the progression of osteoarthritis: an in vitro and in vivo study. J Inflamm (Lond) 2023; 20:14. [PMID: 37055831 PMCID: PMC10100120 DOI: 10.1186/s12950-023-00339-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2023] Open
Abstract
Osteoarthritis (OA) is a common joint disease and is the main cause of physical disability in the elderly. Currently, there is no adequate therapeutic strategy to reverse the progression of OA. Many natural plant extracts have received attention in the treatment of OA due to their potential anti-inflammatory properties, and reduced incidence of adverse events. Dioscin (Dio), a natural steroid saponin, has been demonstrated to inhibit the release of inflammatory cytokines in mouse and rat models of various diseases, and has a protective effect in chronic inflammatory diseases. However, whether Dio alleviates OA progression remains to be explored. In this research, our purposes were to investigate the therapeutic potential of Dio in OA. The results demonstrated that Dio exerted anti-inflammatory effects by repressing NO, PGE2, iNOS and COX-2. Moreover, the application of Dio could repress IL-1β-induced overexpression of matrix metalloproteinases (MMPs, including MMP1, MMP3, and MMP13) and ADAMTS-5, and improve the synthesis of collagen II and aggrecan, which contribute to the maintenance of chondrocyte matrix homeostasis. The underlying mechanism involved the inhibition of the MAPK and NF-κB signaling pathways by Dio. Furthermore, the treatment of Dio significantly improved the pain behaviors of rat OA models. The in vivo study revealed that Dio could ameliorate cartilage erosion and degradation. These results collectively indicate that Dio can be used as a promising and effective agent for the therapy of OA.
Collapse
Affiliation(s)
- Qing Ding
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruizhuo Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianqi Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoze Jing
- Department of Orthopedics, Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, China
| | - Tian Ma
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanxi Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongqi Zhao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wenkai Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Zhao T, Wang S, Liu W, Shen J, Dai Y, Shi M, Huang X, Wei Y, Li T, Zhang X, Xie Z, Wang N, Qin D, Li Z. Clinical efficacy of Yiqi Yangxue formula on knee osteoarthritis and unraveling therapeutic mechanism through plasma metabolites in rats. Front Genet 2023; 14:1096616. [PMID: 37091797 PMCID: PMC10113924 DOI: 10.3389/fgene.2023.1096616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
Objective: To observe the clinical efficacy and safety of Yiqi Yangxue formula (YQYXF) on knee osteoarthritis (KOA), and to explore the underlying therapeutic mechanism of YQYXF through endogenous differential metabolites and their related metabolic pathways.Methods: A total of 61 KOA patients were recruited and divided into the treatment group (YQYXF, 30 cases) and the control group (celecoxib, Cxb, 31 cases). Effects of these two drugs on joint pain, swelling, erythrocyte sedimentation rate (ESR) and c-reactive protein (CRP) were observed, and their safety and adverse reactions were investigated. In animal experiments, 63 SD rats were randomly divided into normal control (NC) group, sham operation (sham) group, model (KOA) group, Cxb group, as well as low-dose (YL), medium-dose (YM), and high-dose groups of YQYXF (YH). The KOA rat model was established using a modified Hulth method. Ultra-high-performance liquid chromatography/Q Exactive HF-X Hybrid Quadrupole-Orbitrap Mass (UHPLC-QE-MS)-based metabolomics technology was used to analyze the changes of metabolites in plasma samples of rats. Comprehensive (VIP) >1 and t-test p < 0.05 conditions were used to screen the disease biomarkers of KOA, and the underlying mechanisms of YQYXF were explored through metabolic pathway enrichment analysis. The related markers of YQYXF were further verified by ELISA (enzyme-linked immunosorbent assay).Results: YQYXF can improve joint pain, swelling, range of motion, joint function, Michel Lequesen index of severity for osteoarthritis (ISOA) score, Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score, ESR, and CRP. No apparent adverse reactions were reported. In addition, YQYXF can improve cartilage damage in KOA rats, reverse the abnormal changes of 16 different metabolites, and exert an anti-KOA effect mainly through five metabolic pathways. The levels of reactive oxygen species (ROS) and glutathione (GSH) were significantly decreased after the treatment of YQYXF.Conclusion: YQYXF can significantly improve the clinical symptoms of KOA patients without obvious adverse reactions. It mainly improved KOA through modulating lipid metabolism-related biomarkers, reducing lipid peroxidation and oxidative stress.
Collapse
Affiliation(s)
- Ting Zhao
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Shiqi Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Wenbin Liu
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Jiayan Shen
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Youwu Dai
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Mingqin Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaoyi Huang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yuanyuan Wei
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Tao Li
- Qujing Hospital Affiliated to Yunnan University of Traditional Chinese Medicine, Qujing, China
| | - Xiaoyu Zhang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaohu Xie
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Na Wang
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
- *Correspondence: Zhaofu Li, ; Na Wang, ; Dongdong Qin,
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
- *Correspondence: Zhaofu Li, ; Na Wang, ; Dongdong Qin,
| | - Zhaofu Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
- *Correspondence: Zhaofu Li, ; Na Wang, ; Dongdong Qin,
| |
Collapse
|
13
|
Lee YM, Kim M, Yuk HJ, Kim SH, Kim DS. Siraitia grosvenorii Residual Extract Inhibits Inflammation in RAW264.7 Macrophages and Attenuates Osteoarthritis Progression in a Rat Model. Nutrients 2023; 15:nu15061417. [PMID: 36986147 PMCID: PMC10058211 DOI: 10.3390/nu15061417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease characterised by cartilage degeneration and chondrocyte inflammation. We investigated the anti-inflammatory effects of the Siraitia grosvenorii residual extract (SGRE) in lipopolysaccharide (LPS)-induced RAW264.7 macrophages in vitro and its anti-osteoarthritic effects in a monosodium iodoacetate (MIA)-induced OA rat model. SGRE dose-dependently decreased nitric oxide (NO) production in LPS-induced RAW264.7 cells. Moreover, SGRE reduced the pro-inflammatory mediator (cyclooxygenase-2 (COX2), inducible NO synthase (iNOS), and prostaglandin E2 (PGE2)) and pro-inflammatory cytokine (interleukin-(IL)-1β, IL-6, and tumour necrosis factor (TNF-α)) levels. SGRE suppressed nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathway activation in RAW264.7 macrophages, thus reducing inflammation. Rats were orally administered SGRE (150 or 200 mg/kg) or the positive control drug JOINS (20 mg/kg) 3 days before MIA injection, and once daily for 21 days thereafter. SGRE elevated the hind paw weight-bearing distribution, thus relieving pain. It also reduced inflammation by inhibiting inflammatory mediator (iNOS, COX-2, 5-LOX, PGE2, and LTB4) and cytokine (IL-1β, IL-6, and TNF-α) expression, downregulating cartilage-degrading enzymes, such as MMP-1, -2, -9, and -13. SGRE significantly reduced the SOX9 and extracellular matrix component (ACAN and COL2A1) levels. Therefore, SGRE is a potential therapeutic active agent against inflammation and OA.
Collapse
Affiliation(s)
- Yun Mi Lee
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (Y.M.L.)
| | - Misun Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (Y.M.L.)
| | - Heung Joo Yuk
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (Y.M.L.)
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, Daejeon 34520, Republic of Korea
| | - Dong-Seon Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea; (Y.M.L.)
- Correspondence: ; Tel.: +82-42-868-9639
| |
Collapse
|
14
|
Stevioside attenuates osteoarthritis via regulating Nrf2/HO-1/NF-κB pathway. J Orthop Translat 2023; 38:190-202. [DOI: 10.1016/j.jot.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
|
15
|
Ding L, Liao T, Yang N, Wei Y, Xing R, Wu P, Li X, Mao J, Wang P. Chrysin ameliorates synovitis and fibrosis of osteoarthritic fibroblast-like synoviocytes in rats through PERK/TXNIP/NLRP3 signaling. Front Pharmacol 2023; 14:1170243. [PMID: 37021049 PMCID: PMC10067567 DOI: 10.3389/fphar.2023.1170243] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
Objective: Synovitis and fibrosis are common pathological features of knee osteoarthritis (KOA). The interaction of synovitis and fibrosis can promote KOA progression. Chrysin (CHR), a natural flavonoid, may treat inflammation and prevent fibrosis. However, the effect and mechanism of CHR in KOA synovitis and fibrosis remains unclear. Methods: The KOA model was established in male SD rats by anterior cruciate ligament transection (ACLT), and histological analysis was used to evaluate synovitis and fibrosis. IL-6, IL-1β and TNF-α mRNA expression in synovial tissue was measured by qRT‒PCR. Immunohistochemistry (IHC) was performed to detect GRP78, ATF-6 and TXNIP expression in vivo. Synovial fibroblasts (SFs) were treated with TGF-β1 to stimulate the inflammatory response and fibrosis. CCK-8 assays were used to detect the viability of CHR-treated SFs. The IL-1β level was detected by immunofluorescence analysis. Coimmunoprecipitation (Co-IP) and double immunofluorescence colocalization were used to detect the physiological interaction between TXNIP and NLRP3. The expression of fibrosis-related mediators and PERK/TXNIP/NLRP3 signaling molecules was detected by western blotting and qRT-PCR. Results: Four weeks after CHR treatment, pathological sections and associated scores showed that CHR improved synovitis and fibrosis in the ACLT model. In vitro, CHR attenuated the TGF-β1-induced inflammatory response and fibrosis in SFs. Moreover, CHR suppressed the expression of synovial fibrosis markers and PERK/TXNIP/NLRP3 signaling molecules in the synovial tissue of rats with ACLT and cultured SFs. More importantly, we found that CHR inhibited TXNIP-NLRP3 interactions in TGF-β-induced SFs. Conclusion: Our findings indicate that CHR can ameliorate synovitis and fibrosis in KOA. The underlying mechanism may be related to the PERK/TXNIP/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Liang Ding
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Taiyang Liao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nan Yang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yibao Wei
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Runlin Xing
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Peng Wu
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaochen Li
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jun Mao
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- *Correspondence: Jun Mao, ; Peimin Wang,
| | - Peimin Wang
- Department of Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Liaoning, China
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
- *Correspondence: Jun Mao, ; Peimin Wang,
| |
Collapse
|
16
|
Liang J, Wang S, Hu J, Hong X, Zhu M, Liu X, Alswadeh M, Mo F, Dai M. Targeted inhibition of TXNRD1 prevents cartilage extracellular matrix degeneration by activating Nrf2 pathway in osteoarthritis. Biochem Biophys Res Commun 2022; 635:267-276. [PMID: 36308906 DOI: 10.1016/j.bbrc.2022.10.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis, a prevalent orthopedic disease, can affect the elderly and causes impairment. The degradation and aberrant homeostasis of cartilage extracellular matrix figure pivotally in the progression of osteoarthritis. Thioredoxin systems plays a role in a wide range of biological processes, including cell proliferation, apoptosis, and oxidative stress. The present study aimed to investigate the unique function and underlying pathophysiological mechanism of TXNRD1 in chondrocytes. An upregulated expression of TXNRD1 was observed in the articular cartilage of osteoarthritis patients compared with normal articular cartilage. Furthermore, in vitro experiments showed that the expression of TXNRD1 was also abnormally increased in IL-1β-induced primary mouse chondrocytes. Silencing TXNRD1 using siRNA in chondrocytes could effectively inhibit the expression of ADAMTS5 and MMP13, and enhance the expression of COL2A1 and SOX9. The same was true for auranofin, an inhibitor of TXNRD1. This phenomenon indicated that inhibition of TXNRD1 attenuated il-1β-induced metabolic imbalance of extracellular matrix (ECM) and the progression of chondrocyte osteoarthritis. Further mechanism analysis revealed that the activation of Nrf2 signaling pathway and the expression of heme oxygenase-1 (HO-1) were increased upon TXNRD1 inhibition. Furthermore, auranofin was found to attenuate DMM-induced osteoarthritis progression in vivo. Therefore, the pharmacological downregulation of TXNRD1 may provide an effective novel therapy for OA.
Collapse
Affiliation(s)
- Jianhui Liang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Song Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Jiawei Hu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xin Hong
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Meisong Zhu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Xuqiang Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Momen Alswadeh
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China
| | - Fengbo Mo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China.
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, China.
| |
Collapse
|
17
|
Cai W, Zhang Y, Jin W, Wei S, Chen J, Zhong C, Zhong Y, Tu C, Peng H. Procyanidin B2 ameliorates the progression of osteoarthritis: An in vitro and in vivo study. Int Immunopharmacol 2022; 113:109336. [DOI: 10.1016/j.intimp.2022.109336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/21/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
18
|
Toropitsyn E, Pravda M, Rebenda D, Ščigalková I, Vrbka M, Velebný V. A composite device for viscosupplementation treatment resistant to degradation by reactive oxygen species and hyaluronidase. J Biomed Mater Res B Appl Biomater 2022; 110:2595-2611. [PMID: 35727166 DOI: 10.1002/jbm.b.35114] [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: 07/29/2021] [Revised: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is one of the most common musculoskeletal disorders in the world. OA is often associated with the loss of viscoelastic and tribological properties of synovial fluid (SF) due to degradation of hyaluronic acid (HA) by reactive oxygen species (ROS) and hyaluronidases. Viscosupplementation is one of the ways how to effectively restore SF functions. However, current viscosupplementation products provide only temporal therapeutic effect because of short biological half-life. In this article we describe a novel device for viscosupplementation (NV) based on the cross-linked tyramine derivative of HA, chondroitin sulfate (CS), and high molecular weight HA by online determination of viscoelastic properties loss during degradation by ROS and hyaluronidase. Rheological and tribological properties of developed viscosupplement were compared with HA solutions with different molecular weights in the range 500-2000 kDa, which are currently commonly used as medical devices for viscosupplementation treatment. Moreover, based on clinical practice and scientific literature all samples were also diluted by model OA SF in the ratio 1:1 (vol/vol) to better predict final properties after injection to the joint. The observed results confirmed that NV exhibits appropriate rheological properties (viscosity, elastic, and viscous moduli) comparable with healthy SF and maintain them during degradation for a significantly longer time than HA solutions with molecular weight in the range 500-2000 kDa and cross-linked material without CS.
Collapse
Affiliation(s)
- Evgeniy Toropitsyn
- Contipro a.s., Dolní Dobrouč, Czech Republic.,Biocev, First Faculty of Medicine Charles University, Vestec, Czech Republic
| | | | - David Rebenda
- Faculty of Mechanical Engineering, Brno University of Technology, Brno, Czech Republic
| | | | - Martin Vrbka
- Faculty of Mechanical Engineering, Brno University of Technology, Brno, Czech Republic
| | | |
Collapse
|
19
|
Ding DF, Xue Y, Wu XC, Zhu ZH, Ding JY, Song YJ, Xu XL, Xu JG. Recent Advances in Reactive Oxygen Species (ROS)-Responsive Polyfunctional Nanosystems 3.0 for the Treatment of Osteoarthritis. J Inflamm Res 2022; 15:5009-5026. [PMID: 36072777 PMCID: PMC9443071 DOI: 10.2147/jir.s373898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/11/2022] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is an inflammatory and degenerative joint disease with severe effects on individuals, society, and the economy that affects millions of elderly people around the world. To date, there are no effective treatments for OA; however, there are some treatments that slow or prevent its progression. Polyfunctional nanosystems have many advantages, such as controlled release, targeted therapy and high loading rate, and have been widely used in OA treatment. Previous mechanistic studies have revealed that inflammation and ROS are interrelated, and a large number of studies have demonstrated that ROS play an important role in different types of OA development. In this review article, we summarize third-generation ROS-sensitive nanomaterials that scavenge excessive ROS from chondrocytes and osteoclasts in vivo. We only focus on polymer-based nanoparticles (NPs) and do not review the effects of drug-loaded or heavy metal NPs. Mounting evidence suggests that polyfunctional nanosystems will be a promising therapeutic strategy in OA therapy due to their unique characteristics of being sensitive to changes in the internal environment.
Collapse
Affiliation(s)
- Dao-Fang Ding
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yan Xue
- Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Centre), Tongji University, Shanghai, People’s Republic of China
| | - Xi-Chen Wu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Zhi-Heng Zhu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jia-Ying Ding
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yong-Jia Song
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, People’s Republic of China
- Correspondence: Xiao-Ling Xu, Shulan International Medical College, Zhejiang Shuren University, 8 Shuren Street, Hangzhou, 310015, People’s Republic of China, Email
| | - Jian-Guang Xu
- Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Jian-Guang Xu, Center of Rehabilitation Medicine, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 200000, People’s Republic of China, Email
| |
Collapse
|
20
|
Wan H, Li C, Yang Y, Chen D. Loganin attenuates interleukin-1 β-induced chondrocyte inflammation, cartilage degeneration, and rat synovial inflammation by regulating TLR4/MyD88/NF-κB. J Int Med Res 2022; 50:3000605221104764. [PMID: 36000146 PMCID: PMC9421229 DOI: 10.1177/03000605221104764] [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] [Indexed: 11/24/2022] Open
Abstract
Objective Inflammation plays a crucial part in osteoarthritis (OA) development. This
work aimed to explore loganin’s role and molecular mechanism in inflammation
and clarify its anti-inflammatory effects in OA treatment. Methods Chondrocytes were stimulated using interleukin (IL)-1β and loganin at two
concentrations (1 μM and 10 μM). Nitric oxide (NO) and prostaglandin E2
(PGE2) expression was assessed. Real-time polymerase chain reaction was used
to evaluate inducible NO synthase (iNOS), cyclooxygenase (COX)-2, IL-6, and
tumor necrosis factor (TNF)-α mRNA levels. Western blot was used to
investigate TLR4, MyD88, p-p65, and IκB-α expression. p65 nuclear
translocation, synovial inflammatory response, and cartilage degeneration
were also assessed. Results Loganin significantly reduced IL-1β-mediated PGE2, NO, iNOS, and COX-2
expression compared with that of the IL-1β stimulation group. The
TLR4/MyD88/NF-κB pathway was suppressed by loganin, which decreased
inflammatory cytokine (TNF-α and IL-6) levels compared with those of the
IL-1β stimulation group. Loganin inhibited IL-1β-mediated NF-κB p65 nuclear
translocation compared with that of the IL-1β stimulation group. Loganin
partially suppressed cartilage degeneration and the synovial inflammatory
response in vivo. Conclusion This work demonstrated that loganin inhibited IL-1β-mediated inflammation in
rat chondrocytes through TLR4/MyD88/NF-κB pathway regulation, thereby
reducing rat cartilage degeneration and the synovial inflammatory
response.
Collapse
Affiliation(s)
- Haishan Wan
- Emergency Trauma Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Chaoyi Li
- Department of Joint Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Yi Yang
- Department of Spinal Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Dingzhong Chen
- Department of Spinal Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| |
Collapse
|
21
|
Luo P, Huang Q, Chen S, Wang Y, Dou H. Asiaticoside ameliorates osteoarthritis progression through activation of Nrf2/HO-1 and inhibition of the NF-κB pathway. Int Immunopharmacol 2022; 108:108864. [PMID: 35623293 DOI: 10.1016/j.intimp.2022.108864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 02/08/2023]
Abstract
Osteoarthritis has become the fourth cause of disability in the world and its occurrence and development are caused by apoptosis and extracellular matrix (ECM) degradation of chondrocytes. Asiaticoside (ASI) is a triterpene saponin compound obtained from Centella Asiatica and has anti-inflammatory and anti-apoptotic effects in various diseases. However, its effects on OA are not clear. In this study, we reported that ASI has a protective effect on the occurrence and progression of OA in vivo and in vitro, and demonstrated its potential molecular mechanism. In vitro, ASI treatment inhibited the release of pro-apoptotic factors induced by TBHP and promoted the release of the anti-apoptotic proteins. In addition, ASI promotes the expression of Aggrecan and Collagen II, while inhibiting the expression of thrombospondin motifs 5 (ADAMTS5) and matrix metalloproteinase-13 (MMP-13), which causes extracellular matrix (ECM) degradation. Mechanistically, ASI exerts its anti-apoptotic effect by activating the Nrf2/HO-1 pathway and preventing p65 from binding to DNA. Similarly, in vivo, ASI has been shown to have a protective effect in a mouse OA model. The conclusion is that our research shows that ASI can be used as a potential drug for the treatment of OA.
Collapse
Affiliation(s)
- Peng Luo
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qishan Huang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Suo Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yinghui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Haicheng Dou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang Province, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
22
|
Signal-On and Highly Sensitive Electrochemiluminescence Biosensor for Hydrogen Sulfide in Joint Fluid Based on Silver-Ion-Mediated Base Pairs and Hybridization Chain Reaction. CHEMOSENSORS 2022. [DOI: 10.3390/chemosensors10070250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Hydrogen sulfide (H2S) in joint fluid acts as a signal molecule to regulate joint inflammation. Direct detection of H2S in joint fluid is of great significance for the diagnosis and treatment of arthritis. However, due to the low volume of joint fluid and low H2S concentration, existing methods face the problem of the insufficient limit of detection. In this study, a highly sensitive biosensor was proposed by designing a primer probe and combining it with hybrid chain reaction (HCR) under the strong interaction between metal ions and H2S to achieve H2S detection. The primer probe containing multiple cytosine (C) sequences was fixed on a gold electrode, and the C–Ag–C hairpin structure was formed under the action of Ag+. In the presence of H2S, it can combine with Ag+ in the hairpin structure to form Ag2S, which leads to the opening of the hairpin structure and triggers the hybridization chain reaction (HCR) with another two hairpin structures (H1 and H2). A large number of double-stranded nucleic acid structures can be obtained on the electrode surface. Finally, Ru(phen)32+ can be embedded into the double chain structure to generate the electrochemiluminescence (ECL) signal. The linear response of the H2S biosensor ranged from 0.1000 to 1500 nM, and the limit of detection concentration of H2S was 0.0398 nM. The developed biosensor was successfully used to determine H2S in joint fluid.
Collapse
|
23
|
The Role of Mitochondrial Metabolism, AMPK-SIRT Mediated Pathway, LncRNA and MicroRNA in Osteoarthritis. Biomedicines 2022; 10:biomedicines10071477. [PMID: 35884782 PMCID: PMC9312479 DOI: 10.3390/biomedicines10071477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease characterized by degeneration of articular cartilage and causes severe joint pain, physical disability, and impaired quality of life. Recently, it was found that mitochondria not only act as a powerhouse of cells that provide energy for cellular metabolism, but are also involved in crucial pathways responsible for maintaining chondrocyte physiology. Therefore, a growing amount of evidence emphasizes that impairment of mitochondrial function is associated with OA pathogenesis; however, the exact mechanism is not well known. Moreover, the AMP-activated protein kinase (AMPK)–Sirtuin (SIRT) signaling pathway, long non-coding RNA (lncRNA), and microRNA (miRNA) are important for regulating the physiological and pathological processes of chondrocytes, indicating that these may be targets for OA treatment. In this review, we first focus on the importance of mitochondria metabolic dysregulation related to OA. Then, we show recent evidence on the AMPK-SIRT mediated pathway associated with OA pathogenesis and potential treatment options. Finally, we discuss current research into the effects of lncRNA and miRNA on OA progression or inhibition.
Collapse
|
24
|
Yuan A, Wu P, Zhong Z, He Z, Li W. Long non-coding RNA Gm37494 alleviates osteoarthritis chondrocyte injury via the microRNA-181a-5p/GABRA1 axis. J Orthop Surg Res 2022; 17:304. [PMID: 35689264 PMCID: PMC9185876 DOI: 10.1186/s13018-022-03202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/25/2022] [Indexed: 11/10/2022] Open
Abstract
Objective This study was conducted to investigate the effect of long non-coding RNA (lncRNA) Gm37494 on osteoarthritis (OA) and its related molecular mechanism. Methods The cartilage tissues were obtained from OA patients, and an OA mouse model was induced by the destabilization of the medial meniscus, followed by measurement of Gm37494, microRNA (miR)-181a-5p, GABRA1 mRNA, and the encoded GABAARα1 protein expression. Thereafter, a cellular model was induced by interleukin-1β (IL-1β) treatment in chondrocytes, followed by ectopic and silencing experiments. Chondrocyte proliferation was detected by CCK-8 and EdU assays, chondrocyte apoptosis by flow cytometry and western blot, and the levels of inflammatory factors by ELISA. The binding of Gm37494 to miR-181a-5p was evaluated by dual-luciferase reporter gene and RIP assays, and that of GABRA1 to miR-181a-5p by dual-luciferase reporter gene and RNA pull-down assays. Results OA patients and mice had decreased GABRA1 mRNA and GABAARα1 protein levels and elevated miR-181a-5p expression in cartilage tissues. Additionally, Gm37494 was poorly expressed in OA mice. Mechanistically, Gm37494 directly bound to and inversely modulated miR-181a-5p that negatively targeted GABRA1. In IL-1β-induced chondrocytes, Gm37494 overexpression enhanced cell proliferation and suppressed cell apoptosis and inflammation, whereas further miR-181a-5p up-regulation or GABRA1 silencing abolished these trends. Conclusions Conclusively, Gm37494 elevated GABRA1 expression by binding to miR-181a-5p, thus ameliorating OA-induced chondrocyte damage. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-022-03202-5.
Collapse
Affiliation(s)
- Aidong Yuan
- Department of Joint and Sports Medicine, The First People's Hospital of Shaoguan City, No.3 Dongdi South Road, Zhenjiang District, Shaoguan, 512000, Guangdong, People's Republic of China.
| | - Penghuan Wu
- Department of Joint and Sports Medicine, The First People's Hospital of Shaoguan City, No.3 Dongdi South Road, Zhenjiang District, Shaoguan, 512000, Guangdong, People's Republic of China
| | - Zhinian Zhong
- Department of Joint and Sports Medicine, The First People's Hospital of Shaoguan City, No.3 Dongdi South Road, Zhenjiang District, Shaoguan, 512000, Guangdong, People's Republic of China
| | - Zhengyan He
- Department of Joint and Sports Medicine, The First People's Hospital of Shaoguan City, No.3 Dongdi South Road, Zhenjiang District, Shaoguan, 512000, Guangdong, People's Republic of China
| | - Wenhu Li
- Department of Joint and Sports Medicine, The First People's Hospital of Shaoguan City, No.3 Dongdi South Road, Zhenjiang District, Shaoguan, 512000, Guangdong, People's Republic of China
| |
Collapse
|
25
|
Dual functions of microRNA-17 in maintaining cartilage homeostasis and protection against osteoarthritis. Nat Commun 2022; 13:2447. [PMID: 35508470 PMCID: PMC9068604 DOI: 10.1038/s41467-022-30119-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/14/2022] [Indexed: 12/16/2022] Open
Abstract
Damaged hyaline cartilage has no capacity for self-healing, making osteoarthritis (OA) "difficult-to-treat". Cartilage destruction is central to OA patho-etiology and is mediated by matrix degrading enzymes. Here we report decreased expression of miR-17 in osteoarthritic chondrocytes and its deficiency contributes to OA progression. Supplementation of exogenous miR-17 or its endogenous induction by growth differentiation factor 5, effectively prevented OA by simultaneously targeting pathological catabolic factors including matrix metallopeptidase-3/13 (MMP3/13), aggrecanase-2 (ADAMTS5), and nitric oxide synthase-2 (NOS2). Single-cell RNA sequencing of hyaline cartilage revealed two distinct superficial chondrocyte populations (C1/C2). C1 expressed physiological catabolic factors including MMP2, and C2 carries synovial features, together with C3 in the middle zone. MiR-17 is highly expressed in both superficial and middle chondrocytes under physiological conditions, and maintains the physiological catabolic and anabolic balance potentially by restricting HIF-1α signaling. Together, this study identified dual functions of miR-17 in maintaining cartilage homeostasis and prevention of OA.
Collapse
|
26
|
Hu W, Mao C, Sheng W. The protective effect of kirenol in osteoarthritis: an in vitro and in vivo study. J Orthop Surg Res 2022; 17:195. [PMID: 35365162 PMCID: PMC8974005 DOI: 10.1186/s13018-022-03063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a chronic degenerative disease, its main characteristic involves articular cartilage destruction and inflammation response, absent of effective medical treatment. Our current research aimed to explore anti-inflammatory effect of kirenol, a diterpenoid natural product compound, in the development of OA and its potential molecular mechanism through in vitro and in vivo study. METHODS In vitro, chondrocytes were pretreated with kirenol for 2 h before IL-1β stimulation. Production of NO, PGE2, TNF-α, IL-6, aggrecan, collagen-II, MMP13and ADAMTS5 were evaluated by the Griess reaction and ELISAs. The mRNA (aggrecan and collagen-II) and protein expression (COX-2, iNOS, P65, IκB, PI3K, AKT) were measured by qRT-PCR and Western blot respectively. Immunofluorescence was used to assess the expression of collagen-II and P65. The in vivo effect of kirenol was evaluated in mice OA models induced by destabilization of the medial meniscus (DMM). RESULTS We found that kirenol inhibited IL-1β-induced expression of NO, PGE2, TNF-α, IL-6, COX-2, iNOS, ADAMTS-5. Besides, kirenol remarkably decreased IL-1β-induced degradation of aggrecan and collagen-II. Furthermore, kirenol significantly inhibited IL-1β-induced phosphorylation of PI3K/Akt and NF-κB signaling. In vivo, the cartilage in kirenol-treated mice exhibited less cartilage degradation and lower OARSI scores. CONCLUSIONS Taken together, the results of this study provide potent evidence that kirenol could be utilized as a potentially therapeutic agent in prevention and treatment of OA.
Collapse
Affiliation(s)
- Wei Hu
- Department of Spine Surgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Chao Mao
- Department of Spine Surgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Weibin Sheng
- Department of Spine Surgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China.
| |
Collapse
|
27
|
Elmholt SB, Hede KC, Christensen BB, Thomsen JS, Lind M. The Effect of Bone Marrow Stimulation for Cartilage Repair on the Subchondral Bone Plate. Cartilage 2022; 13:19476035221074011. [PMID: 35098739 PMCID: PMC9137303 DOI: 10.1177/19476035221074011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To investigate the effect of bone-marrow stimulation (BMS) on subchondral bone plate morphology and remodeling compared to untreated subchondral bone in a validated minipig model. METHODS Three Göttingen minipigs received BMS with drilling as treatment for two chondral defects in each knee. The animals were euthanized after six months. Follow-up consisted of a histological semiquantitative evaluation using a novel subchondral bone scoring system and micro computed tomography (µCT) of the BMS subchondral bone. The histological and microstructural properties of the BMS-treated subchondral bone were compared to that of the adjacent healthy subchondral bone. RESULTS The µCT analysis showed that subchondral bone treated with BMS had significantly higher connectivity density compared to adjacent untreated subchondral bone (26 1/mm3 vs. 21 1/mm3, P = 0.048). This was the only microstructural parameter showing a significant difference. The histological semiquantitative score differed significantly between the subchondral bone treated with BMS and the adjacent untreated subchondral (8.0 vs. 10 P = < 0.001). Surface irregularities were seen in 43% and bone overgrowth in 27% of the histological sections. Only sparse formation of bone cysts was detected (1%). CONCLUSIONS BMS with drilling does not cause extensive changes to the subchondral bone microarchitecture. Furthermore, the morphology of BMS subchondral bone resembled that of untreated subchondral bone with almost no formation of bone cyst, but some surface irregularities and bone overgrowth.
Collapse
Affiliation(s)
- Simone Birkebæk Elmholt
- Department of Orthopaedics, Aarhus University Hospital, Aarhus, Denmark,Simone Birkebæk Elmholt, Department of Orthopaedics, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark.
| | | | | | | | - Martin Lind
- Department of Orthopaedics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
28
|
Li X, Zhou W, Wang L, Ye Y, Li T. Transcranial Direct Current Stimulation Alleviates the Chronic Pain of Osteoarthritis by Modulating NMDA Receptors in Midbrain Periaqueductal Gray in Rats. J Pain Res 2022; 15:203-214. [PMID: 35115824 PMCID: PMC8801364 DOI: 10.2147/jpr.s333454] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022] Open
Abstract
Purpose Osteoarthritis (OA) is the most common cause to lead to chronic pain. Transcranial direct current stimulation (tDCS) has been widely used to treat nerve disorders and chronic pain. The benefits of tDCS for chronic pain are apparent, but its analgesic mechanism is still unclear. This study observed the analgesic effects of tDCS on OA-induced chronic pain and the changes of NMDA receptor levels in PAG after tDCS treatment in rats to explore the analgesic mechanism of tDCS. Methods After establishing chronic pain by injecting monosodium iodoacetate (MIA) into the rat ankle joint, the rats received tDCS for 14 consecutive days (20 min/day). Before tDCS treatment, Ifenprodil (the selective antagonist of NMDAR2B) was given to rats in different ways: intracerebroventricular (i.c.v.) injection or intraperitoneal (i.p.) injection. The Von Frey and hot plate tests were applied to assess the pain-related behaviors at different time points. The expression level of NMDAR2B was evaluated in midbrain periaqueductal gray (PAG) by Western blot. In addition, NMDAR2B and c-Fos were observed by the Immunohistochemistry staining after tDCS treatment. Results The mechanical allodynia and thermal hyperalgesia were produced after MIA injection. However, tDCS treatment reverted the mechanical allodynia and thermal hyperalgesia. Moreover, tDCS treatment significantly increased the expression of NMDAR2B and the proportion of positive stained cells of NMDAR2B. Besides that, the tDCS treatment also decreased the proportion of positive stained cells of c-Fos in PAG. However, these changes did not occur in the rats given the Ifenprodil (i.c.v.). Conclusion These results indicate that tDCS may increase the expression of NMDA receptors in PAG and strengthen the NMDA receptors-mediated antinociception to alleviate OA-induced chronic pain in rats.
Collapse
Affiliation(s)
- Xinhe Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Wenwen Zhou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Lin Wang
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Yinshuang Ye
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Tieshan Li
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
- Correspondence: Tieshan Li, Email
| |
Collapse
|
29
|
Duan J, Shen T, Dong H, Han S, Li G. Association of the Expression Levels of Long-Chain Noncoding RNA TUG1 and Its Gene Polymorphisms with Knee Osteoarthritis. Genet Test Mol Biomarkers 2021; 25:102-110. [PMID: 33596137 DOI: 10.1089/gtmb.2020.0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objective: To study the association of the expression levels of long noncoding RNA Taurine-upregulated gene 1 (lncRNA TUG1) and TUG1 polymorphisms with knee osteoarthritis (KOA). Materials and Methods: A total of 255 KOA patients and 255 controls from May 2017 to December 2019 were selected for the study. Sanger sequencing was conducted to detect the genotypes of the TUG1 rs5749201, rs7284767, and rs886471 loci in all study subjects. Unconditional logistic regression analysis was used to calculate odds ratios and 95% confidence intervals, and the associations between the TUG1 rs574901, rs7284767 and rs886471 loci and KOA risk were analyzed. Multifactor dimensionality reduction was used to analyze the interactions among alleles at the three TUG1 loci examined. Quantitative real-time polymerase chain reaction was used to evaluate the expression levels of TUG1 lncRNA in plasma. Results: A total of 255 KOA patients and 255 control subjects completed the study. After adjusting for the factors of gender, age, body mass index, smoking history, drinking history, and family history, we found that the carriers of the A allele of the TUG1 rs5749201 locus were 1.36 times more likely to develop KOA than the carriers of the T allele (95% confidence interval [CI] = 1.05-1.75, p = 0.02); the G allele of the rs7284767 locus was a protective factor for KOA (odds ratio [OR] = 0.71, 95% CI = 0.54-0.92, p = 0.01); and the allelic variation at rs886471 G > T led to an increased risk of KOA by 2.34 times (95% CI = 1.53-3.57, p < 0.01). We also found that the GAG haplotype for the three loci was significantly associated with the increased risk of KOA (OR = 2.77, 95% CI = 1.67-4.57, p < 0.01). There was no correlation found between the TUG1 rs886471, rs5749201, and rs7284767 single nucleotide polymorphisms loci and the severity of KOA. The allelic variation at TUG1 rs5749201 T > A, rs886471 T > G were associated with decreased levels of TUG1 lncRNA in the plasma of the subjects, while the allelic variation at rs7284767 A > G was associated with increased levels of TUG1 lncRNA in plasma (p = 0.01, p < 0.01, p < 0.01). Conclusion: Plasma TUG1 lncRNA levels and loci at the TUG1 rs5749201, rs7284767, and rs886471 loci are associated with KOA risk.
Collapse
Affiliation(s)
- Jiqiang Duan
- Department of Orthopedics, Zibo Central Hospital, Zibo, China
| | - Tiehui Shen
- Department of Orthopedics, Zibo Central Hospital, Zibo, China
| | - Hao Dong
- Department of Orthopedics, Zibo Central Hospital, Zibo, China
| | - Shiliang Han
- Department of Orthopedics, Zibo Central Hospital, Zibo, China
| | - Guo Li
- Department of Orthopedics, Chengdu First People's Hospital, Chengdu, China
| |
Collapse
|
30
|
Yuan T, Wang X, Cai D, Liao M, Liu R, Qin J. Anti-arthritic and cartilage damage prevention via regulation of Nrf2/HO-1 signaling by glabridin on osteoarthritis. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
31
|
Zhu DC, Wang YH, Lin JH, Miao ZM, Xu JJ, Wu YS. Maltol inhibits the progression of osteoarthritis via the nuclear factor-erythroid 2-related factor-2/heme oxygenase-1 signal pathway in vitro and in vivo. Food Funct 2021; 12:1327-1337. [PMID: 33443518 DOI: 10.1039/d0fo02325f] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Osteoarthritis (OA) is a common degenerative joint disease characterized by articular cartilage degeneration and inflammation. Currently, there is hardly any effective treatment for OA due to its complicated pathology and the severe side effects of the treatment drugs used. It has been reported that maltol, a Maillard reaction product derived from ginseng, inhibits inflammation and oxidative stress in several animal models. However, the potential anti-inflammatory effects of maltol in OA treatment are unknown. This study aimed to evaluate the anti-inflammatory effects of maltol on interleukin (IL)-1β-induced mouse chondrocytes and protective effects of maltol on these chondrocytes in medial meniscus destabilization (DMM) OA mouse models. Mice, randomly divided into maltol (n = 15), vehicle (n = 15) and control (n = 15) groups were treated with the same dose of maltol or saline, respectively. The cartilage tissues were extracted for histological analysis 8 weeks postoperative. For the in vitro studies, chondrocytes were treated with 10 ng mL-1 IL-1β combined with maltol at different concentrations. In vitro assays showed that the maltol pre-treatment significantly inhibited the expressions of multiple inflammatory factors induced by IL-1β, such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), nitric oxide (NO), interleukin-6 (IL-6) and tumor necrosis factor (TNF-α). In addition, maltol alleviated the degradation of the extracellular matrix (ECM) by inhibiting the expressions of matrix metalloproteinase-13 (MMP13) and thrombospondin motif 5 (ADAMTS5), as well as reversing the degradation of aggrecan and collagen II. Moreover, maltol suppressed nuclear factor kappa B (NF-κB) signaling by activating the nuclear factor-erythroid 2-related factor-2 (Nrf2) in in vitro and in vivo studies. These findings indicate that maltol reduces the inflammation induced by IL-1β in chondrocytes. Therefore, the results of this study indicated that maltol may be a potential drug for the effective treatment of OA.
Collapse
Affiliation(s)
- Ding-Chao Zhu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China. and Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China and The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yi-Han Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China. and Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China and The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jia-Hao Lin
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China. and Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China and The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhi-Min Miao
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China. and Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China and The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jia-Jing Xu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China. and Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China and The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yao-Sen Wu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China. and Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China and The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
32
|
Wang X, Li Z, Cui Y, Cui X, Chen C, Wang Z. Exosomes Isolated From Bone Marrow Mesenchymal Stem Cells Exert a Protective Effect on Osteoarthritis via lncRNA LYRM4-AS1-GRPR-miR-6515-5p. Front Cell Dev Biol 2021; 9:644380. [PMID: 34124036 PMCID: PMC8193855 DOI: 10.3389/fcell.2021.644380] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Objectives The aim of this study was to investigate the effects of exosomes isolated from human bone marrow mesenchymal stem cells (BMSCs) on osteoarthritis (OA) and a competitive endogenous RNA (ceRNA) network. Methods Exosomes were isolated from human BMSCs and characterized by transmission electron microscopy (TEM), Nanosight (NTA), and western blotting. Chondrocytes were treated with interleukin-1β (IL-1β) and then transfected with exosomes. Cell viability and apoptosis were determined using Cell Counting Kit-8 (CCK-8) and flow cytometry, respectively. Cells with IL-1β and exosomes were sequenced, and differentially expressed lncRNAs (DE-lncRNAs) and miRNAs (DE-miRNAs) were identified. Thereafter, a ceRNA network (LYRM4-AS1-GRPR-miR-6515-5p) was chosen for further validation. Results TEM, NTA, and western blotting showed that exosomes were successfully isolated, and PKH67 staining showed that exosomes could be taken up by IL-1β-induced chondrocytes. Compared with the control group, IL-1β significantly decreased cell viability and promoted apoptosis (P < 0.05), while exosomes reversed the changes induced by IL-1β. For MMP3, AKT, and GRPR, IL-1β upregulated their expression, while exosomes downregulated their expression. For PTEN, there was no significant difference in PTEN expression between the control and IL-1β groups; however, exosomes markedly upregulated PTEN expression. By sequencing, 907 DE-lncRNAs and 25 DE-miRNAs were identified, and a ceRNA network was constructed. The dual-luciferase reporter gene indicated that LYRM4-AS1, miR-6515-5, and GRPR interacted with each other. The results of cell experiments showed that LYRM4-AS1 regulated the growth of IL-1β-induced chondrocytes by GRPR/miR-6515-5p. Conclusion Exosomes may alleviate OA inflammation by regulating the LYRM4-AS1/GRPR/miR-6515-5p signaling pathway.
Collapse
Affiliation(s)
- Xiuhui Wang
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Zhuokai Li
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yin Cui
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xu Cui
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Cheng Chen
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Zhou S, Shi J, Wen H, Xie W, Han X, Li H. A chondroprotective effect of moracin on IL-1β-induced primary rat chondrocytes and an osteoarthritis rat model through Nrf2/HO-1 and NF-κB axes. Food Funct 2021; 11:7935-7945. [PMID: 32832965 DOI: 10.1039/d0fo01496f] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Osteoarthritis (OA) is a common joint disease characterized by cartilage degeneration and inflammation. Although moracin is known to play a role in anti-inflammation and anti-oxidation in several inflammatory diseases, its anti-inflammatory effect on OA remains largely unknown. Therefore, in order to explore the role of moracin in OA, we investigated the anti-inflammatory effect of moracin on interleukin (IL)-β-induced rat chondrocytes in vitro and surgically induced OA rat models in vivo. Rat chondrocytes were pretreated using moracin (0, 5, 10, 15 μmol L-1) and then stimulated with IL-β (10 ng ml-1). Results showed that moracin reduced the expression of IL-1β-induced nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor-α (TNF-α), IL-6, inducible nitric oxide synthase (iNOS), and cyclooxygenase (COX)-2 in both rat chondrocytes and cell culture supernatants. Besides, IL-1β-induced degradation of aggrecan and collage II, and the high expression of matrix metalloproteinase-13 (MMP-13) and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS)-5 were also reversed by moracin. Moreover, moracin inhibited the translocation of p65 from the cytoplasm to nucleus induced by IL-1β and activated the Nrf2/HO-1 signaling pathway in chondrocytes. In OA rat models, moracin prevented cartilage of rats from destruction. All these findings above indicated that moracin could be a potentially effective drug for treating OA.
Collapse
Affiliation(s)
- Siqi Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jiaqi Shi
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441021, China
| | - Haiyan Wen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xie
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Xiaotao Han
- Wuhan National High Magnetic Field Center, Huazhong University of Science and Technology, Wuhan, 430074, China and State Key Laboratory of Advanced Electromagnetic Engineering and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Haohuan Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
34
|
Li Y, Chen M, Yan J, Zhou W, Gao S, Liu S, Li Q, Zheng Y, Cheng Y, Guo Q. Tannic acid/Sr 2+-coated silk/graphene oxide-based meniscus scaffold with anti-inflammatory and anti-ROS functions for cartilage protection and delaying osteoarthritis. Acta Biomater 2021; 126:119-131. [PMID: 33684536 DOI: 10.1016/j.actbio.2021.02.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/16/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023]
Abstract
Tissue engineering method provides a promising solution for meniscus repair and regeneration. However, the inflammatory environment that persists after meniscus injury in the knee joint impedes meniscus tissue regeneration. The purpose of this study was to investigate the applicability of silk/graphene oxide (GO)-based meniscus scaffold modified with tannic acid (TA)/Sr2+ coating for the elimination of inflammatory cytokines and reactive oxygen species (ROS) under osteoarthritis (OA) environment along with cartilage protection by using a rat model. The self-assembled coating composed of a series of TA-Sr2+ complex concentrations was formed by a facile, rapid, and efficient method on the scaffold. The phenolic hydroxyl groups on the coating endowed the meniscus scaffold with excellent anti-inflammatory and ROS scavenging capacities. We also found that the coating could promote cell migration in a mock wound model and could increase extracellular matrix secretion in vitro. Moreover, the coating components at a certain concentration played an effective role in delaying OA and providing cartilage protection in the rat model. The expression of inflammation cytokines (e.g., IL-6, IL-8, and MMPs) in rat knee tissue was significantly downregulated, and cartilage degeneration and OA damage were also inhibited according to tissue staining results and the OARSI (Osteoarthritis Research Society International) scoring system. Combining these performances, we suggest that this silk/GO-based scaffold modified with TA/Sr2+ coating could have broader application prospects by virtue of its effective and user-friendly properties. STATEMENT OF SIGNIFICANCE: The biological properties of the meniscus play a role in activating and regulating the metabolic and inflammatory responses that influence the homeostasis of joint health and ultimately lead to knee osteoarthritis (OA). The inflammation condition of the knee joint may exacerbate the degeneration of meniscus and cartilage. The present study aimed to develop a functional coating composed of tannic acid/Sr2+ complex on a silk/graphene oxide-based meniscus scaffold and to endow the scaffold with anti-inflammatory and ROS elimination capacities during the meniscus regeneration process to protect cartilage and delay OA development. The in vitro cytocompatibility study and the in vivo rat OA model study revealed that the coating was effective in promoting cell migration, facilitating ECM secretion, inhibiting inflammation, and delaying OA development.
Collapse
Affiliation(s)
- Yangyang Li
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Mingxue Chen
- Department of Orthopedic Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, No. 31 Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Jianglong Yan
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Wenhao Zhou
- Shaanxi Key Laboratory of biomedical metal materials, Northwest Institute for Non-ferrous Metal Research, Xi'an 710016, China
| | - Shuang Gao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuyun Liu
- Institute of Orthopedics, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries in PLA, Chinese PLA General Hospital, Beijing 100853, China
| | - Qiyao Li
- Department of Biomedical Engineering, Materials Research Institute, Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Yan Cheng
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| | - Quanyi Guo
- Institute of Orthopedics, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Lab of Musculoskeletal Trauma & War Injuries in PLA, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
35
|
Lu H, Wang W, Kang X, Lin Z, Pan J, Cheng S, Zhang J. Hydrogen (H 2) Alleviates Osteoarthritis by Inhibiting Apoptosis and Inflammation via the JNK Signaling Pathway. J Inflamm Res 2021; 14:1387-1402. [PMID: 33880054 PMCID: PMC8053515 DOI: 10.2147/jir.s297622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/18/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a very common condition and leads to joint pain, disability, and price tag all over the world. Pathogenesis of OA is closely related to numerous inflammatory and apoptosis cytokines. Hydrogen (H2) reportedly exhibits a diversity of effects such as anti-apoptotic, anti-inflammatory, and anti-oxidative properties via the JNK pathway. However, it is unknown whether H2 has a protective effect against OA via the JNK signaling pathway. Therefore, the aim of this study was to figure out whether hydrogen has protective effect on chondrocyte and further explore the possible underlying mechanism. METHODS The chondrocytes were obtained from the human cartilage tissues. Cells were stimulated by TBHP and treated with hydrogen. In vitro treatment effects were evaluated by Western blot assay, real-time PCR, immunofluorescence and TUNEL method. We conducted mice model of destabilization of the medial meniscus (DMM) and treated with hydrogen. In vivo treatment effects were evaluated by X-ray imaging assay, safranin O (SO) staining, TUNEL staining and immunohistochemical assay. RESULTS Our results showed that hydrogen can inhibit inflammatory factors (ADAMTS5 and MMP13) and apoptosis factors (cleaved caspase-3, cytochrome c, and Bax) in TBHP-induced chondrocytes. Furthermore, hydrogen can suppress the activation of JNK signaling pathway, whereas the effect of hydrogen can be abolished by anisomycin (a JNK activator). In vivo results showed that hydrogen can down-regulate the expression of p-JNK and cleaved caspase-3 expression. CONCLUSION We uncovered that hydrogen (H2) could alleviate apoptosis response and ECM degradation in human chondrocytes via inhibiting the activation of the JNK signaling pathway. Meanwhile, in the surgically-induced DMM mice model, treatment with hydrogen (H2) performed a significant role in OA progression.
Collapse
Affiliation(s)
- Hongwei Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Wei Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Xiaodiao Kang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Zeng Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Jun Pan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, People’s Republic of China
| | - Shaowen Cheng
- Trauma Center, First Affiliated Hospital of Hainan Medical University, Haikou, People’s Republic of China
| | - Jingdong Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People’s Republic of China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, People’s Republic of China
- Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, People’s Republic of China
| |
Collapse
|
36
|
Fioravanti A, Tenti S, Cheleschi S. MiR-214-3p, a novel possible therapeutic target for the pathogenesis of osteoarthritis. EBioMedicine 2021; 66:103300. [PMID: 33774326 PMCID: PMC8024903 DOI: 10.1016/j.ebiom.2021.103300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/10/2021] [Indexed: 10/26/2022] Open
Affiliation(s)
- Antonella Fioravanti
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, Azienda Ospedaliera Universitaria Senese, Policlinico Le Scotte, 53100 Siena, Italy
| | - Sara Tenti
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, Azienda Ospedaliera Universitaria Senese, Policlinico Le Scotte, 53100 Siena, Italy
| | - Sara Cheleschi
- Department of Medicine, Surgery and Neuroscience, Rheumatology Unit, Azienda Ospedaliera Universitaria Senese, Policlinico Le Scotte, 53100 Siena, Italy.
| |
Collapse
|
37
|
Lu H, Fu C, Kong S, Wang X, Sun L, Lin Z, Luo P, Jin H. Maltol prevents the progression of osteoarthritis by targeting PI3K/Akt/NF-κB pathway: In vitro and in vivo studies. J Cell Mol Med 2021; 25:499-509. [PMID: 33211383 PMCID: PMC7810946 DOI: 10.1111/jcmm.16104] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 08/15/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022] Open
Abstract
Osteoarthritis (OA), a prevalent degenerative arthritis disease, principle characterized by the destruction of cartilage and associated with the inflammatory response. Maltol, a product formed during the processing of red ginseng (Panax ginseng, CA Meyer), has been reported to have the potential effect of anti-inflammatory. However, its specific mechanisms are not demonstrated. We investigated the protective effect of maltol in the progression of OA both in vitro and in vivo experiments. Human chondrocytes were pre-treated with maltol (0, 20, 40, 60 μM, 24 hours) and incubated with IL-1β (10 ng/mL, 24 hours) in vitro. Expression of PGE2, TNF-α and NO was measured by the ELISA and Griess reaction. The expression of iNOs, COX-2, aggrecan, ADAMTS-5, MMP-13, IκB-α, p65, P-AKT, AKT, PI3K and P-PI3K was analysed by Western blotting. The expression of collagen II and p65-active protein was detected by immunofluorescence. Moreover, the serious level of OA was evaluated by histological analysis in vivo. We identified that maltol could suppress the IL-1β-stimulated generation of PGE2 and NO. Besides, maltol not only suppressed the production of COX-2, iNOs, TNF-α, IL-6, ADAMTS-5, MMP-13, but also attenuated the degradation of collagen II and aggrecan. Furthermore, maltol remarkably suppressed the phosphorylation of PI3K/AKT and NF-κB induced by IL-1β in human OA chondrocytes. Moreover, maltol could block the cartilage destroy in OA mice in vivo. To date, all data indicate maltol is a potential therapeutic agent by inhibiting inflammatory response via the regulation of NF-κB signalling for OA.
Collapse
Affiliation(s)
- Hongwei Lu
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| | - Changchang Fu
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
- Department of NeonatologyThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Suyan Kong
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| | - Xudong Wang
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| | - Lin Sun
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| | - Zeng Lin
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| | - Peng Luo
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| | - Haidong Jin
- Department of OrthopaedicsThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouChina
- The Second School of MedicineWenzhou Medical UniversityWenzhouChina
- The Key Orthopaedic Laboratory of Zhejiang ProvinceBone Research InstituteWenzhouChina
| |
Collapse
|
38
|
Hamza RZ, Al-Salmi FA, El-Shenawy NS. Chitosan and Lecithin Ameliorate Osteoarthritis Symptoms Induced by Monoiodoacetate in a Rat Model. Molecules 2020; 25:molecules25235738. [PMID: 33291821 PMCID: PMC7730914 DOI: 10.3390/molecules25235738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/28/2020] [Accepted: 11/28/2020] [Indexed: 01/08/2023] Open
Abstract
The present work aimed to assess the chondroprotective influence of chitosan and lecithin in a monoiodoacetate (MIA)-induced experimental osteoarthritis (OA) model. Forty male rats weighing 180–200 g were randomly distributed among the following five experimental groups (eight per group): control, MIA-induced OA, MIA-induced OA + chitosan, MIA-induced OA + lecithin, and MIA-induced OA + chitosan + lecithin. The levels of TNF-α, IL6, RF, ROS, and CRP, as well as mitochondrial markers such as mitochondrial swelling, cytochrome C oxidase (complex IV), MMP, and serum oxidative/antioxidant status (MDA level) (MPO and XO activities) were elevated in MIA-induced OA. Also, SDH (complex II) activity in addition to the levels of ATP, glutathione (GSH), and thiol was markedly diminished in the MIA-induced OA group compared to in control rats. These findings show that mitochondrial function is associated with OA pathophysiology and suggest that chitosan and lecithin could be promising potential ameliorative agents in OA animal models. Lecithin was more effective than chitosan in ameliorating all of the abovementioned parameters.
Collapse
Affiliation(s)
- Reham Z. Hamza
- Department of Biology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Department of Zoology, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
- Correspondence: ; Tel.: +96-6531-355470 or +20-111-8500-586
| | - Fawziah A. Al-Salmi
- Department of Biology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Nahla S. El-Shenawy
- Department of Zoology, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
39
|
Mao X, Fu P, Wang L, Xiang C. Mitochondria: Potential Targets for Osteoarthritis. Front Med (Lausanne) 2020; 7:581402. [PMID: 33324661 PMCID: PMC7726420 DOI: 10.3389/fmed.2020.581402] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a common and disabling joint disorder that is mainly characterized by cartilage degeneration and narrow joint spaces. The role of mitochondrial dysfunction in promoting the development of OA has gained much attention. Targeting endogenous molecules to improve mitochondrial function is a potential treatment for OA. Moreover, research on exogenous drugs to improve mitochondrial function in OA based on endogenous molecular targets has been accomplished. In addition, stem cells and exosomes have been deeply researched in the context of cartilage regeneration, and these factors both reverse mitochondrial dysfunctions. Thus, we hypothesize that biomedical approaches will be applied to the treatment of OA. Furthermore, we have summarized the global status of mitochondria and osteoarthritis research in the past two decades, which will contribute to the research field and the development of novel treatment strategies for OA.
Collapse
Affiliation(s)
- Xingjia Mao
- Department of Orthopedic, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Panfeng Fu
- Department of Respiratory and Critical Care, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Linlin Wang
- Department of Basic Medicine Sciences, The School of Medicine of Zhejiang University, Hangzhou, China
| | - Chuan Xiang
- Department of Orthopedic, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
40
|
Chen G, Song Y, Ma F, Ma Y. Anti-arthritic activity of D-carvone against complete Freund's adjuvant-induced arthritis in rats through modulation of inflammatory cytokines. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:453-462. [PMID: 33093267 PMCID: PMC7585598 DOI: 10.4196/kjpp.2020.24.6.453] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 05/20/2020] [Accepted: 08/11/2020] [Indexed: 11/15/2022]
Abstract
Chronic joint pain due to loss of cartilage function, degradation of subchondral bone, and related conditions are common plights of an arthritis patient. Antioxidant compounds could solve the problems in arthritic condition. The objective of this study was to evaluate the anti-arthritic activity of D-carvone against complete Freund's adjuvant (CFA)-induced arthritis in rats. D-carvone was orally administered for 25 days at the doses of 30 and 60 mg/kg against CFA-induced arthritic rats. Changes in body weight, paw swelling, organ index, hematological parameters, oxidative stress markers, inflammatory cytokines, and histopathology were recorded. Oral treatment of D-carvone significantly improved the body weight, reduced the paw swelling, edema formation, and organ index in arthritic rats. The levels of white blood cells were reduced, red blood cells and hemoglobin levels were improved in D-carvone treated arthritic rats. Lipid peroxidation levels were lowered whereas enzymatic and non-enzymatic antioxidants were significantly elevated by D-carvone administration against arthritic rats. D-carvone significantly modulated inflammatory cytokine levels and improved the ankle joint pathology against CFA-induced arthritic inflammation. In conclusion, D-carvone proved significant anti-arthritic activity against CFA-induced arthritis in rats.
Collapse
Affiliation(s)
- Guifang Chen
- Rheumatism and Immunology Ward of Integrated Traditional Chinese and Western Medicine, Jinan, Shandong Province 250013, P. R. China
| | - Yuxiu Song
- Department of Dermatology Pain Management, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250013, P. R. China
| | - Fang Ma
- Department of Rheumatology and Immunology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250013, P. R. China
| | - Yuxia Ma
- Department of Rheumatology and Immunology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250013, P. R. China
| |
Collapse
|
41
|
Kriston-Pál É, Haracska L, Cooper P, Kiss-Tóth E, Szukacsov V, Monostori É. A Regenerative Approach to Canine Osteoarthritis Using Allogeneic, Adipose-Derived Mesenchymal Stem Cells. Safety Results of a Long-Term Follow-Up. Front Vet Sci 2020; 7:510. [PMID: 32903517 PMCID: PMC7438407 DOI: 10.3389/fvets.2020.00510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/03/2020] [Indexed: 01/11/2023] Open
Abstract
Mesenchymal stem cells (MSC) are emerging as an effective therapeutic tool in treating canine osteoarthritis (OA). In this report, we focused on the questions of whether MSC transplantation has long-term beneficial effects for the improvement in motion and also evaluated the safety of MSC injection. Visceral adipose tissue, a surgical waste obtained during routine ovariectomy served as a source of allogeneic MSCs and used to treat OA. Altogether, fifty-eight dogs were transplanted in the study suffering from OA in the elbow (42 animals), hip (5), knee (8), ankle (2), and hock (1). The effect of MSC transplantation was evaluated by the degree of lameness at a 4-5-years follow-up period based on the owners' subjective observations. The results showed that 83% of the OA patients improved or retained improvement in lameness. Clinical safety of the treatment was assessed by evaluating the coincidence of tumors or other diseases and other adverse reactions (such as local inflammation) after MSC cell therapy. Two incidences of local inflammation for <1 week at the site of injection were reported. No other adverse reactions were detected post-treatment. Sixteen dogs died during the study, 4 due to cancer and 12 due to other diseases, diagnosed by veterinarians. Overall, our survey suggests that MSC transplantation has long-term beneficial effects in reducing lameness. Moreover, no enrichment in a specific cause of death was observed in the transplanted animals, compared to reported literature. Our data suggest that MSC treatment could be an effective and safe long-term therapy for canine OA.
Collapse
Affiliation(s)
| | - Lajos Haracska
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - Paul Cooper
- Assentra Limited, Chelmsford, United Kingdom
| | - Endre Kiss-Tóth
- University of Sheffield, Department of Infection, Immunity & Cardiovascular Disease, Sheffield, United Kingdom
| | | | - Éva Monostori
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| |
Collapse
|
42
|
Refat MS, Hamza RZ, Adam A, Saad HA, Gobouri AA, Al-Salmi FA, Altalhi T, El-Megharbel SM. Synthesis of N, N'-bis(1,5-dimethyl-2-phenyl-1,2-dihydro-3-oxopyrazol-4-yl) sebacamide that ameliorate osteoarthritis symptoms and improve bone marrow matrix structure and cartilage alterations induced by monoiodoacetate in the rat model: "Suggested potent anti-inflammatory agent against COVID-19". Hum Exp Toxicol 2020; 40:325-341. [PMID: 32840387 PMCID: PMC7447864 DOI: 10.1177/0960327120945779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To assess the chondroprotective effect and influence of
N,N′-bis(1,5-dimethyl-2-phenyl-1,2-dihydro-3-oxopyrazol-4-yl)
sebacamide (dpdo) that was synthesized through the reaction of phenazone with
sebacoyl chloride and screened for its biological activity especially as
anti-arthritic and anti-inflammatory agent in a monoiodoacetate (MA)-induced
experimental osteoarthritis (OA) model. Thirty male albino rats weighing
“190–200 g” were divided randomly into three groups (10 each): control,
MA-induced OA, and MA-induced OA + dpdo. In MA-induced OA rat, the tumor
necrosis factor alpha, interleukin 6, C-reactive protein, rheumatoid factors,
reactive oxygen species, as well as all the mitochondrial markers such as
mitochondria membrane potential, swelling mitochondria, cytochrome
c oxidase (complex IV), and serum oxidative/antioxidant
status (malondialdehyde level and activities of myeloperoxidase and xanthine
oxidase) are elevated. Also, the activity of succinate dehydrogenase (complex
II), levels of ATP, the level of glutathione (GSH), and thiol were markedly
diminished in the MA-induced OA group compared to the normal control rats. These
findings showed that mitochondrial function is associated with OA
pathophysiological alterations and high gene expressions of (IL-6, TNF-a, and
IL-1b) and suggests a promising use of dpdo as potential ameliorative agents in
the animal model of OA and could act as anti-inflammatory agent in case of
severe infection with COVID-19. It is clearly appeared in improving the bone
cortex and bone marrow in the treated group with the novel compound in
histological and transmission electron microscopic sections which is a very
important issue today in fighting severe infections that have significant
effects on the blood indices and declining of blood corpuscles like COVID-19, in
addition to declining the genotoxicity and inflammation induced by MA in male
rats. The novel synthesized compound was highly effective in improving all the
above mentioned parameters.
Collapse
Affiliation(s)
- M S Refat
- Department of Chemistry, Faculty of Science, 125895Taif University, Taif, Saudi Arabia.,Department of Chemistry, Faculty of Science, Port Said University, Port Said, Egypt
| | - R Z Hamza
- Department of Biology, Faculty of Science, 125895Taif University, Taif, Saudi Arabia.,Department of Zoology, Faculty of Science, 68799Zagazig University, Zagazig, Egypt
| | - Ama Adam
- Department of Chemistry, Faculty of Science, 125895Taif University, Taif, Saudi Arabia
| | - H A Saad
- Department of Chemistry, Faculty of Science, 125895Taif University, Taif, Saudi Arabia.,Department of Chemistry, Faculty of Science, 68799Zagazig University, Zagazig, Egypt
| | - A A Gobouri
- Department of Chemistry, Faculty of Science, 125895Taif University, Taif, Saudi Arabia
| | - F A Al-Salmi
- Department of Biology, Faculty of Science, 125895Taif University, Taif, Saudi Arabia
| | - T Altalhi
- Department of Chemistry, Faculty of Science, 125895Taif University, Taif, Saudi Arabia
| | - S M El-Megharbel
- Department of Chemistry, Faculty of Science, 125895Taif University, Taif, Saudi Arabia.,Department of Chemistry, Faculty of Science, 68799Zagazig University, Zagazig, Egypt
| |
Collapse
|
43
|
Wang H, Li J, Cheng Y, Yao J. Association of Long-Chain Noncoding RNA H19 and MEG3 Gene Polymorphisms and Their Interaction with Risk of Osteoarthritis in a Chinese Han Population. Genet Test Mol Biomarkers 2020; 24:328-337. [PMID: 32364812 DOI: 10.1089/gtmb.2019.0230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Huang Wang
- Department of Orthopedics, Hangzhou Dingqiao Hospital (Hangzhou Hospital of Traditional Chinese Medicine, Dingqiao District), Hangzhou, China
| | - Jian Li
- Department of Spinal Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Ye Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Yao
- Department of Orthopedics, Hangzhou Dingqiao Hospital (Hangzhou Hospital of Traditional Chinese Medicine, Dingqiao District), Hangzhou, China
| |
Collapse
|
44
|
Biochemical Signals Mediate the Crosstalk between Cartilage and Bone in Osteoarthritis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5720360. [PMID: 32337258 PMCID: PMC7165323 DOI: 10.1155/2020/5720360] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/30/2019] [Accepted: 03/26/2020] [Indexed: 12/12/2022]
Abstract
Osteochondral junction is a functional unit comprising the articular cartilage, calcified cartilage, and subchondral bone. Alteration in any component of this composite unit can disrupt the joint integrity and function directly or indirectly. Biochemical signals mediate the crosstalk between tissues and play an essential role in the initiation and progression of osteoarthritis. As osteoarthritis progresses, abnormal subchondral bone remodelling leads to increased angiogenesis and porosity of the subchondral bone plate, which further triggers biochemical signals to mediate the crosstalk between cartilage and bone, contributing to the progression of osteoarthritis. Notably, common biochemical signals include the TGF-β/Smad, Wnt/β-catenin, RANK/RANKL/OPG, and MAPK pathways. This biomarker crosstalk network is the basis of osteoarthritis pathogenesis, and some of their key regulators may be potential therapeutic targets for osteoarthritis drug therapy. This review summarised the biochemical crosstalk between cartilage and bone in the pathogenesis of osteoarthritis, which may provide the basis for the discovery of osteoarthritis treatment targets.
Collapse
|
45
|
Lin Z, Lin C, Fu C, Lu H, Jin H, Chen Q, Pan J. The protective effect of Ellagic acid (EA) in osteoarthritis: An in vitro and in vivo study. Biomed Pharmacother 2020; 125:109845. [PMID: 32058211 DOI: 10.1016/j.biopha.2020.109845] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/27/2019] [Accepted: 12/29/2019] [Indexed: 01/11/2023] Open
Abstract
Osteoarthritis (OA), a progressive joint disorder, is principally characterized by the degeneration and destruction of the articular cartilage. Ellagic acid (EA), a natural polyphenol found in berries and nuts has shown potent anti-inflammatory effects, however, its effects and underlying mechanisms on OA have seldom been systematically illuminated. In this study, we reported the anti-inflammatory effects of Ellagic acid (EA) in the progression of OA in both in vitro and in vivo experiments. in vitro study, IL-1β-induced expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), Nitric oxide (NO), tumor necrosis factor-alpha (TNF-α), prostaglandin E2 (PGE2), and interleukin-6 (IL-6) were inhibited by Ellagic acid (EA). Moreover, Ellagic acid (EA) down-regulated the IL-1β-stimulated matrix metalloproteinase-13 (MMP-13) and thrombospondin motifs 5 (ADAMTS-5) while up-regulated the collagen of type II and aggrecan. Mechanistically, we revealed that Ellagic acid (EA) suppressed nuclear factor kappa B (NF-κB) signaling in IL-1β -induced chondrocytes. And Ellagic acid (EA)-induced protectiveness in OA development was also shown by the DMM model. Taken together, our data indicate that Ellagic acid (EA) may serve as a potential drug for OA treatment.
Collapse
Affiliation(s)
- Zeng Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xue Yuan Xi Road, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Chen Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xue Yuan Xi Road, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Changchang Fu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Hongwei Lu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xue Yuan Xi Road, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Haidong Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xue Yuan Xi Road, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China
| | - Qin Chen
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Jun Pan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, 109 Xue Yuan Xi Road, Wenzhou, Zhejiang 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China; Bone Research Institute, The Key Orthopaedic Laboratory of Zhejiang Province, Wenzhou, China.
| |
Collapse
|
46
|
Ng J, Little CB, Woods S, Whittle S, Lee FY, Gronthos S, Mukherjee S, Hunter DJ, Worthley DL. Stem cell-directed therapies for osteoarthritis: The promise and the practice. Stem Cells 2020; 38:477-486. [PMID: 31837053 DOI: 10.1002/stem.3139] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is a disease of an entire synovial joint characterized by clinical symptoms and distortion of joint tissues, including cartilage, muscles, ligaments, and bone. Although OA is a disease of all joint tissues, it is a defined accessible compartment and is thus amenable to topical surgical and regenerative therapies, including stem cells. All tissues arise from stem progenitor cells, and the relative capacity of different cellular compartments, and different individuals, to renew tissues into adulthood may be important in the onset of many different degenerative diseases. OA is driven by both mechanical and inflammatory factors, but how these factors affect the proliferation and differentiation of cells into cartilage in vivo is largely unknown. Indeed, our very basic understanding of the physiological cellular kinetics and biology of the stem-progenitor cell unit of the articular cartilage, and how this is influenced by mechano-inflammatory injury, is largely unknown. OA seems, rather deceptively, to be the low-hanging fruit for stem cell therapy. Without the basic understanding of the stem cell and progenitor unit that generate and maintain articular cartilage in vivo, we will continue to waste opportunities to both prevent and manage this disease. In this review, we discuss the biology of chondrogenesis, the stem cell populations that support articular cartilage in health and disease, and future opportunities afforded through the translation of basic articular chondrocyte stem cell biology into new clinical therapies.
Collapse
Affiliation(s)
- Jia Ng
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Christopher B Little
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, New South Wales, Australia.,Raymond Purves Bone & Joint Research Laboratories, Kolling Institute, St. Leonards, Sydney, New South Wales, Australia
| | - Susan Woods
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Samuel Whittle
- Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Francis Y Lee
- Rheumatology Department, Royal North Shore Hospital, Institute of Bone and Joint Research, Kolling Institute, University of Sydney, St. Leonards, New South Wales, Australia
| | - Stan Gronthos
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Mesenchymal Stem Cell Laboratory, University of Adelaide, Adelaide, South Australia, Australia
| | - Siddhartha Mukherjee
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York
| | - David J Hunter
- Northern Clinical School, University of Sydney, St. Leonards, Sydney, New South Wales, Australia
| | - Daniel L Worthley
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
47
|
Zhang Z, Wang X, Li P, Bai M, Qi W. Evaluation of antiarthritic activity of ginkgolic acid against Freund’s adjuvant induced arthritic rat model. BRAZ J PHARM SCI 2020. [DOI: 10.1590/s2175-97902019000418638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Ziqiang Zhang
- Shaanxi Baoji Traditional Chinese Medical Hospital, People’s Republic of China
| | - Xiaogang Wang
- Shaanxi Baoji Traditional Chinese Medical Hospital, People’s Republic of China
| | - Pengshan Li
- Shaanxi Baoji Traditional Chinese Medical Hospital, People’s Republic of China
| | - Minghua Bai
- Shaanxi Baoji Traditional Chinese Medical Hospital, People’s Republic of China
| | - Wenbing Qi
- Shaanxi Baoji Traditional Chinese Medical Hospital, People’s Republic of China
| |
Collapse
|
48
|
Liu X, Wang L, Ma C, Wang G, Zhang Y, Sun S. Exosomes derived from platelet-rich plasma present a novel potential in alleviating knee osteoarthritis by promoting proliferation and inhibiting apoptosis of chondrocyte via Wnt/β-catenin signaling pathway. J Orthop Surg Res 2019; 14:470. [PMID: 31888697 PMCID: PMC6936129 DOI: 10.1186/s13018-019-1529-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 12/19/2019] [Indexed: 03/20/2023] Open
Abstract
Background Platelet-rich plasma (PRP) provides a nonsurgical approach for treating osteoarthritis (OA). Exosomes that play vital roles in intercellular communication have been studied extensively. Here, we investigated the therapeutic potential and molecular mechanism of exosomes derived from PRP (PRP-Exos) in alleviating OA. Methods Exosomes derived from PRP(PRP-Exos) were isolated and purified using the exoEasy Maxi Kit and then identified and analyzed. Primary rabbit chondrocytes were isolated and treated with interleukin 1 beta (IL-1β) to establish the OA model in vitro. Proliferation, migration, and apoptosis assays were measured and compared between PRP-Exos and activated PRP (PRP-As) to evaluate the therapeutic effects on OA. The mechanism involving the Wnt/β-catenin signaling pathway was investigated by Western blot analysis. In vivo, we established animal knee OA model by surgery to compare the therapeutic effect of PRP-Exos and PRP-As. Results We successfully isolated and purified exosomes from PRP using the exoEasy Maxi Kit. We also isolated and identified chondrocytes from the New Zealand white rabbit and established the IL-1β-induced OA model; meanwhile, PRP-Exos and PRP-As both inhibited the release of tumor necrosis factor-α(TNF-α) and there was no statistically significant difference between the two. In proliferation, migration, scratch assay, the promoting effect of PRP-Exos was significantly more better than PRP-As. Furthermore, PRP-Exos could significantly decreased apoptotic rate of OA chondrocyte compared with PRP-As. In Western blot analysis, the expression of β-catenin, and RUNX2, Wnt5a were increased in IL-1β-treated chondrocytes, but PRP-Exos and PRP-As could both reverse these changes, and the reversal effect of the former was better than the latter. In vivo, we found that both PRP-Exos and PRP-As displayed the progression of OA, and the effect of PRP-Exos was obviously better than PRP-As by chondrocyte count and Osteoarthritis Research Society International (OARSI) scoring system. Conclusion The therapeutic effects of PRP-Exos on OA were similar or better compared with those of PRP-As in vitro or in vivo. PRP-Exos acting as carriers containing growth factors derived from PRP present a novel therapy for OA by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xuchang Liu
- Department of Emergency Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.,School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Taian, 271016, Shandong, China
| | - Lubo Wang
- Department of Trauma Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Chengshan Ma
- Department of Emergency Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China
| | - Guozong Wang
- School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China
| | - Yuanji Zhang
- School of Medicine, Shandong University, 44 Wenhua Road, Jinan, 250012, Shandong, China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
49
|
Huang X, You Y, Xi Y, Ni B, Chu X, Zhang R, You H. p-Coumaric Acid Attenuates IL-1β-Induced Inflammatory Responses and Cellular Senescence in Rat Chondrocytes. Inflammation 2019; 43:619-628. [DOI: 10.1007/s10753-019-01142-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
50
|
Xu Z, Yang H, Zhou X, Li J, Jiang L, Li D, Wu L, Huang Y, Xu N. Genetic variants in mTOR-pathway-related genes contribute to osteoarthritis susceptibility. Int Immunopharmacol 2019; 77:105960. [DOI: 10.1016/j.intimp.2019.105960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 12/23/2022]
|