1
|
Uehara Y, Suzukawa M, Horie M, Igarashi S, Minegishi M, Takada K, Saito A, Nagase H. ZFP36 family expression is suppressed by Th2 cells in asthma, leading to enhanced synthesis of inflammatory cytokines and cell surface molecules. Cell Immunol 2024; 403-404:104859. [PMID: 39067169 DOI: 10.1016/j.cellimm.2024.104859] [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: 04/11/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Asthma is a chronic inflammatory airway disease, in which inflammatory cytokines play a pivotal role. The zinc finger binding protein 36 (ZFP36) family includes ZFP36, ZFP36L1, and ZFP36L2 and is among the RNA-binding proteins (RBPs) reported to cause inflammation. The present study aimed to clarify the roles of the ZFP36 family in asthma, particularly highlighting the relationship between the ZFP36 family and Th2 cells, which are key players in type 2 inflammation in asthma. Real-time PCR analysis revealed the preferential expression of ZFP36 family mRNAs in human white blood cells. Gene expression analysis using public datasets from the GEO database (https://www.ncbi.nlm.nih.gov/gds) showed significantly suppressed expression of ZFP36 family mRNAs in patients with asthma compared to that in healthy controls. Using multiple cytokine assays, Th2 cell transfection with ZFP36 family siRNAs enhanced the expression of inflammatory cytokines IL-8, IFN-γ, CCL3/MIP-1α, CCL4/MIP-1β, and TNF-α and cell surface molecules CCR4 (CD194) and PSGL-1 (CD162). Treatment with IL-2, 4, and 15 significantly suppressed, and corticosteroid significantly enhanced the expressions of ZFP36 family mRNAs by Th2 cells. In conclusion, the ZFP36 family expressed by Th2 cells was suppressed in patients with asthma, leading to the enhanced expression of cytokines and cell surface molecules. Suppressed ZFP36 expression in asthma may be involved in the enhancement of airway inflammation, and the ZFP36 family may be a therapeutic target for inflammatory diseases, including asthma.
Collapse
Affiliation(s)
- Yuki Uehara
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo, 204-8585, Japan; Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| | - Maho Suzukawa
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo, 204-8585, Japan.
| | - Masafumi Horie
- Department of Molecular and Cellular Pathology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Sayaka Igarashi
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo, 204-8585, Japan
| | - Masaaki Minegishi
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo, 204-8585, Japan
| | - Kazufumi Takada
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, 3-1-1 Takeoka, Kiyose-City, Tokyo, 204-8585, Japan; Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, 113-8655, Japan
| | - Akira Saito
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Nagase
- Division of Respiratory Medicine and Allergology, Department of Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-Ku, Tokyo, 173-8605, Japan
| |
Collapse
|
2
|
Lee Y, Kim HE, Kwak JS, Park CS, Chun JS. The cereblon-AMPK (AMP-activated protein kinase) axis in chondrocytes regulates the pathogenesis of osteoarthritis. Osteoarthritis Cartilage 2024:S1063-4584(24)01364-5. [PMID: 39218203 DOI: 10.1016/j.joca.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE AMP-activated protein kinase (AMPK) dysregulation is implicated in osteoarthritis (OA), but the mechanisms underlying this dysregulation remain unclear. We investigated the role of cereblon, a substrate-recognition protein within the E3-ligase ubiquitin complex, in AMPK dysregulation and OA pathogenesis. METHODS Cereblon expression was examined in human (n = 5) and mouse (n = 10) OA cartilage. The role of cereblon was investigated through its adenoviral overexpression (n = 10) or knockout (KO, n = 15) in the destabilization of the medial meniscus (DMM)-operated mice. The therapeutic potentials of the chemical cereblon degrader, TD-165, and the AMPK activator, metformin, were assessed through intra-articular (IA) injection to mice (n = 15). RESULTS Immunostaining revealed that cereblon is upregulated in human and mouse OA cartilage. In DMM model mice, cartilage destruction was exacerbated by overexpression of cereblon in mouse joint tissues (OARSI grade; 1.11 [95% CI: 0.50 to 2.75]), but inhibited in global (-2.50 [95% CI: -3.00 to -1.17]) and chondrocyte-specific (-2.17 [95% CI: -3.14 to -1.06]) cereblon KO mice. The inhibitory effects were more pronounced in mice fed a high-fat diet compared to a regular diet. The degradation of cereblon through IA injection of TD-165 inhibited OA cartilage destruction (-2.47 [95% CI: -3.22 to -1.56]). Mechanistically, cereblon exerts its catabolic effects by negatively modulating AMPK activity within chondrocytes. Consistently, activation of AMPK by IA injection of metformin inhibited posttraumatic OA cartilage destruction (-1.20 ([95% CI: -1.89 to -0.45]). CONCLUSIONS The cereblon-AMPK axis acts as a catabolic regulator of OA pathogenesis and seems to be a promising therapeutic target in animal models of OA.
Collapse
Affiliation(s)
- Yeon Lee
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyo-Eun Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ji-Sun Kwak
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chul-Seung Park
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jang-Soo Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
3
|
Farooq HMU, Yang L, Cao M, Chen Z, Qian A, Dang K. Recent Progress in the Research on RNA-Binding Proteins in Bone Development and Diseases. Int J Mol Sci 2024; 25:7735. [PMID: 39062974 PMCID: PMC11276800 DOI: 10.3390/ijms25147735] [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: 05/22/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
RNA-binding proteins (RBPs), which regulate gene expression through post-transcriptional modifications of RNAs, play a role in diverse biological processes that include bone cell development and bone tissue formation. RBP dysregulation may result in aberrant bone homeostasis and contribute to various bone diseases. The function of RBPs in bone physiology and pathophysiology and the underlying molecular mechanisms have been extensively studied in recent years. This article provides a review of such studies, highlighting the potential of RBPs as pivotal targets for therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | | | - Airong Qian
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (H.M.U.F.); (L.Y.); (Z.C.)
| | - Kai Dang
- Laboratory for Bone Metabolism, Xi’an Key Laboratory of Special Medicine and Health Engineering, Key Laboratory for Space Biosciences and Biotechnology, Research Center for Special Medicine and Health Systems Engineering, NPU-UAB Joint Laboratory for Bone Metabolism, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China; (H.M.U.F.); (L.Y.); (Z.C.)
| |
Collapse
|
4
|
Cha R, Nakagawa S, Arai Y, Inoue A, Okubo N, Fujii Y, Kaihara K, Nakamura K, Kishida T, Mazda O, Takahashi K. Enhancing Cartilage Metabolism in Rats through a Novel Thermal Stimulation Technique with Photosensitizers. Int J Mol Sci 2024; 25:6728. [PMID: 38928434 PMCID: PMC11204344 DOI: 10.3390/ijms25126728] [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: 05/17/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Although the moderate thermal stimulation of articular cartilage exerts chondroprotective effects, it is difficult to effectively heat deep articular cartilage with conventional methods. Photosensitizers increase the ambient temperature using near-infrared (NIR) radiation, which has high tissue permeability. We hypothesized that the intra-articular administration of photosensitizers and NIR irradiation would exert a greater heating effect on articular cartilage. We aimed to evaluate the heating effect of this method on cultured chondrocytes and rat knee cartilage. In vitro, we irradiated a photosensitizer-containing medium with NIR and measured changes in the medium temperature, cytotoxicity, and gene expression of heat shock protein (HSP) 70 and aggrecan (ACAN). In vivo, the knee joints of rats treated with photosensitizers were irradiated with NIR, and changes in intra-articular temperature and gene expression were measured, alongside histological analysis. The results showed that the medium and intra-articular temperature were raised to approximately 40 °C with no apparent disruption to articular cartilage or the immunohistochemically enhanced staining of HSP70 in chondrocytes. The gene expression of HSP70 and ACAN was increased in both cultured and articular cartilage. In summary, this method can safely heat joints and enhance cartilage metabolism by inducing HSP70 expression in articular cartilage. It presents a new hyperthermia therapy with effective cartilage protection.
Collapse
Affiliation(s)
- Ryota Cha
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| | - Shuji Nakagawa
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yuji Arai
- Department of Sports and Para-Sports Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Atsuo Inoue
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| | - Naoki Okubo
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| | - Yuta Fujii
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| | - Kenta Kaihara
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| | - Kei Nakamura
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kenji Takahashi
- Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan; (R.C.)
| |
Collapse
|
5
|
Liu C, Sun Y, Li D, Wang F, Wang H, An S, Sun S. A multifunctional nanogel encapsulating layered double hydroxide for enhanced osteoarthritis treatment via protection of chondrocytes and ECM. Mater Today Bio 2024; 26:101034. [PMID: 38596826 PMCID: PMC11002310 DOI: 10.1016/j.mtbio.2024.101034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/20/2024] [Accepted: 03/19/2024] [Indexed: 04/11/2024] Open
Abstract
Osteoarthritis (OA) is characterized by progressive and irreversible damage to the articular cartilage and a consecutive inflammatory response. However, the majority of clinical drugs for OA treatment only alleviate symptoms without addressing the fundamental pathology. To mitigate this issue, we developed an inflammation-responsive carrier and encapsulated bioactive material, namely, LDH@TAGel. The LDH@TAGel was designed with anti-inflammatory and antioxidative abilities, aiming to directly address the pathology of cartilage damage. In particular, LDH was confirmed to restore the ECM secretion function of damaged chondrocytes and attenuate the expression of catabolic matrix metalloproteinases (Mmps). While TAGel showed antioxidant properties by scavenging ROS directly. In vitro evaluation revealed that the LDH@TAGel could protect chondrocytes from inflammation-induced oxidative stress and apoptosis via the Nrf2/Keap1 system and Pi3k-Akt pathway. In vivo experiments demonstrated that the LDH@TAGel could alleviated the degeneration and degradation of cartilage induced by anterior cruciate ligament transection (ACLT). The OARSI scores indicating OA severity decreased significantly after three weeks of intervention. Moreover, the IVIS image revealed that LDH@TAGel enhances the controlled release of LDH in a manner that can be customized according to the severity of OA, allowing adaptive, precise treatment. In summary, this novel design effectively alleviates the underlying pathological causes of OA-related cartilage damage and has emerged as a promising biomaterial for adaptive, cause-targeted OA therapies.
Collapse
Affiliation(s)
- Changxing Liu
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Yawei Sun
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Dengju Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Fan Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
| | - Senbo An
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250012, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| |
Collapse
|
6
|
Xie Y, Shao F, Ji Y, Feng D, Wang L, Huang Z, Wu S, Sun F, Jiang H, Miyamoto A, Wang H, Zhang C. Network Analysis of Osteoarthritis Progression Using a Steiner Minimal Tree Algorithm. J Inflamm Res 2024; 17:3201-3209. [PMID: 38779430 PMCID: PMC11110812 DOI: 10.2147/jir.s438407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/09/2023] [Indexed: 05/25/2024] Open
Abstract
Purpose To provide a comprehensive analysis of associated genes with osteoarthritis (OA). Here, we reported a network analysis of OA progression by using a Steiner minimal tree algorithm. Methods We collected the OA-related genes through screening the publications in MEDLINE. We performed functional analysis to analyze the associated biochemical pathways of the OA-related genes. Pathway crosstalk analysis was constructed to explore interactions of the enriched pathways. Steiner minimal tree algorithm was used to analyze molecular pathway networks. The average clustering coefficient was compared with the corresponding values of the Osteoarthritis-specific network. The new finding RNA was compared with former single-cell RNA-seq analysis results. Results A gene set with 177 members reported to be significantly associated with Osteoarthritis was collected from 187 studies. Functional enrichment analysis revealed a specific related-OA gene including skeletal system development, cytokine-mediated signaling pathway, inflammatory response, cartilage development, and extracellular matrix organization. We performed a pathway crosstalk analysis among the 72 significantly enriched pathways. A total of 151 of the 177 genes in the Osteoarthritis gene set were included in the human interactome network. There were 31 genes in the former single-cell RNA-seq analysis results. The CLU, ENO1, SRRM1, UBC, HMGB1, NR3C1, NOTCH2NL, and CBX5 have significantly increased expression in seven molecularly defined populations of OA cartilage. Conclusion The Steiner tree-based approach finds new biological molecules associated with OA genes.
Collapse
Affiliation(s)
- Yujie Xie
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Fanglin Shao
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Yuxiu Ji
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Dechao Feng
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Ling Wang
- School of Information and Communication Engineering, University of Electronic Science and Technology, Chengdu, Sichuan, People’s Republic of China
| | - Zonghai Huang
- School of Information and Communication Engineering, University of Electronic Science and Technology, Chengdu, Sichuan, People’s Republic of China
| | - Shengjian Wu
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Fuhua Sun
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| | - Hong Jiang
- Rehabilitation Medicine Department, Xichong County People’s hospital, Nanchong, Sichuan, People’s Republic of China
| | - Akira Miyamoto
- Faculty of Rehabilitation, Nishikyushu University, Kansaitama, Japan
| | - Haiming Wang
- Department of Rehabilitation Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People’s Republic of China
| | - Chi Zhang
- Rehabilitation Medicine Department, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
7
|
Li H, Cui Y, Wang J, Zhang W, Chen Y, Zhao J. Identification and validation of biomarkers related to lipid metabolism in osteoarthritis based on machine learning algorithms. Lipids Health Dis 2024; 23:111. [PMID: 38637751 PMCID: PMC11025229 DOI: 10.1186/s12944-024-02073-5] [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: 01/29/2024] [Accepted: 03/07/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Osteoarthritis and lipid metabolism are strongly associated, although the precise targets and regulatory mechanisms are unknown. METHODS Osteoarthritis gene expression profiles were acquired from the GEO database, while lipid metabolism-related genes (LMRGs) were sourced from the MigSB database. An intersection was conducted between these datasets to extract gene expression for subsequent differential analysis. Following this, functional analyses were performed on the differentially expressed genes (DEGs). Subsequently, machine learning was applied to identify hub genes associated with lipid metabolism in osteoarthritis. Immune-infiltration analysis was performed using CIBERSORT, and external datasets were employed to validate the expression of these hub genes. RESULTS Nine DEGs associated with lipid metabolism in osteoarthritis were identified. UGCG and ESYT1, which are hub genes involved in lipid metabolism in osteoarthritis, were identified through the utilization of three machine learning algorithms. Analysis of the validation dataset revealed downregulation of UGCG in the experimental group compared to the normal group and upregulation of ESYT1 in the experimental group compared to the normal group. CONCLUSIONS UGCG and ESYT1 were considered as hub LMRGs in the development of osteoarthritis, which were regarded as candidate diagnostic markers. The effects are worth expected in the early diagnosis and treatment of osteoarthritis.
Collapse
Affiliation(s)
- Hang Li
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Yubao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, 214023, Jiangsu, China
| | - Jian Wang
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Wei Zhang
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Yuhao Chen
- Wuxi Medical Center, Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China
| | - Jijun Zhao
- Department of Orthopedic, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299 Qing Yang Road, Wuxi, Jiangsu, 214023, China.
| |
Collapse
|
8
|
Jin G, Xu W, Tang H, Cui Y, Zhang H. Bisdemethoxycurcumin, a curcumin, protects chondrocytes, and reduces cartilage inflammation via the NRF2/HO-1/NLRP3 pathway. Immun Inflamm Dis 2024; 12:e1195. [PMID: 38411358 PMCID: PMC10898200 DOI: 10.1002/iid3.1195] [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: 09/26/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND The objective of this thesis is to evaluate the effect of bisdemethoxycurcumin (BDMC) on osteoarthritis (OA) and comprehensively evaluate the role of the Nuclear Factor erythroid 2-Related Factor 2 (Nrf2) signalling pathway in chondrocytes. METHOD In our study, we treated chondrocytes with BDMC in an in vitro chondrocyte assay and measured its influence on extracellular matrix (ECM) expression, downstream heme oxygenase-1 (HO-1) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) levels. RESULTS Our study indicates that BDMC significantly activates the Nrf2 signaling pathway in chondrocytes in vitro. Furthermore, the expression of matrix metalloproteinase 3, interleukin 1β, recombinant a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)4 and (ADAMTS)5 was significantly suppressed by BDMC. CONCLUSION This study confirms the potential for BDMC to activate the Nrf2/HO-1/NLRP3 signalling pathway and alleviate OA symptoms. Therefore, BDMC is a promising therapeutic agent for OA that offers new insights and treatment methods.
Collapse
Affiliation(s)
- Gang Jin
- Department of OrthopedicsTaizhou Hospital Affiliated to Wenzhou Medical University, LinhaiZhejiangChina
| | - Wei Xu
- Department of OrthopedicsTaizhou Hospital Affiliated to Wenzhou Medical University, LinhaiZhejiangChina
| | - Huilin Tang
- Department of OrthopedicsTaizhou Hospital Affiliated to Wenzhou Medical University, LinhaiZhejiangChina
| | - Yaying Cui
- Department of OrthopedicsTaizhou Hospital Affiliated to Wenzhou Medical University, LinhaiZhejiangChina
| | - Han Zhang
- Department of OrthopedicsTaizhou Hospital Affiliated to Wenzhou Medical University, LinhaiZhejiangChina
| |
Collapse
|
9
|
Suminda GGD, Min Y, Ha MW, Ghosh M, Lee DS, Son YO. In vitro and in vivo investigations on arsenic-induced cartilage degeneration in osteoarthritis. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132570. [PMID: 37742380 DOI: 10.1016/j.jhazmat.2023.132570] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/26/2023]
Abstract
Heavy metals found in the environment, including arsenic (As) pose significant risks to human health and present a risk factor for osteoarthritis (OA). This study researched the impact of As on cartilage degeneration by focusing on the role of As in causing OA in mice. We employed chemical inhibition and inductively coupled plasma mass spectrometry analyses to identify the effect of As on chondrocytes as well as studying its accumulation in organs after oral administration in mice. Additionally, the study examined the effect of intra-articular As treatment on the levels of crucial catabolic factors, namely Hif-2α (Epas1) and Zip8 (Slc39a8), during OA progression. Mice that were administered As orally in conjunction with surgically induced joint instability, had heightened cartilage destruction compared to wild-type mice. Quantitative analysis revealed a significant increase in Hif-2α and Zip8 mRNA expression (p = 0.0352,0.0004 respectively) and protein expression (p = 0.0101,0.008 respectively) post oral administration. Our findings illustrated the role of As in influencing crucial cellular functions that are triggered by reactive oxygen species. These events consequently activate the Akt/Hif-2α/NF-κB pathways, leading to disruptions in articular cartilage homeostasis. This study provides a comprehensive understanding of the impact of As on the development of osteoarthritis.
Collapse
Affiliation(s)
| | - Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Min Woo Ha
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Republic of Korea; Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Mrinmoy Ghosh
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences Jeju National University, Jeju-si 63243, Republic of Korea; Department of Biotechnology, School of Bio, Chemical and Processing Engineering (SBCE), Kalasalin-gam Academy of Research and Educational, Krishnankoil 626126, India
| | - Dong-Sun Lee
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Republic of Korea; Health Materials Core-Facility Center, Jeju National University, Jeju-si 63243, Republic of Korea; Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Republic of Korea; Practical Translational Research Center, Jeju National University, Jeju 63243, Republic of Korea.
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju-si 63243, Republic of Korea; Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences Jeju National University, Jeju-si 63243, Republic of Korea; Health Materials Core-Facility Center, Jeju National University, Jeju-si 63243, Republic of Korea; Practical Translational Research Center, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
10
|
Kim D, Heo Y, Kim M, Suminda GGD, Manzoor U, Min Y, Kim M, Yang J, Park Y, Zhao Y, Ghosh M, Son YO. Inhibitory effects of Acanthopanax sessiliflorus Harms extract on the etiology of rheumatoid arthritis in a collagen-induced arthritis mouse model. Arthritis Res Ther 2024; 26:11. [PMID: 38167214 PMCID: PMC10763440 DOI: 10.1186/s13075-023-03241-1] [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: 06/16/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The biological function of Acanthopanax sessiliflorus Harm (ASH) has been investigated on various diseases; however, the effects of ASH on arthritis have not been investigated so far. This study investigates the effects of ASH on rheumatoid arthritis (RA). METHODS Supercritical carbon dioxide (CO2) was used for ASH extract preparation, and its primary components, pimaric and kaurenoic acids, were identified using gas chromatography-mass spectrometer (GC-MS). Collagenase-induced arthritis (CIA) was used as the RA model, and primary cultures of articular chondrocytes were used to examine the inhibitory effects of ASH extract on arthritis in three synovial joints: ankle, sole, and knee. RESULTS Pimaric and kaurenoic acids attenuated pro-inflammatory cytokine-mediated increase in the catabolic factors and retrieved pro-inflammatory cytokine-mediated decrease in related anabolic factors in vitro; however, they did not affect pro-inflammatory cytokine (IL-1β, TNF-α, and IL-6)-mediated cytotoxicity. ASH effectively inhibited cartilage degradation in the knee, ankle, and toe in the CIA model and decreased pannus development in the knee. Immunohistochemistry demonstrated that ASH mostly inhibited the IL-6-mediated matrix metalloproteinase. Gene Ontology and pathway studies bridge major gaps in the literature and provide insights into the pathophysiology and in-depth mechanisms of RA-like joint degeneration. CONCLUSIONS To the best of our knowledge, this is the first study to conduct extensive research on the efficacy of ASH extract in inhibiting the pathogenesis of RA. However, additional animal models and clinical studies are required to validate this hypothesis.
Collapse
Affiliation(s)
- Dahye Kim
- Division of Animal Genetics and Bioinformatics, National Institute of Animal Science, RDA, Wanju, Republic of Korea
| | - Yunji Heo
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
| | - Mangeun Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
| | - Godagama Gamaarachchige Dinesh Suminda
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
| | - Umar Manzoor
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
- Laboratory of Immune and Inflammatory Disease, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
| | - Minhye Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
| | - Jiwon Yang
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
| | - Youngjun Park
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea
- Laboratory of Immune and Inflammatory Disease, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yaping Zhao
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Mrinmoy Ghosh
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea.
- Department of Biotechnology, School of Bio, Chemical and Processing Engineering (SBCE), Kalasalingam Academy of Research and Education, Krishnankoil, Srivilliputhur, 626126, India.
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea.
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City, Jeju Special Self-Governing Province, 63243, Republic of Korea.
- Practical Translational Research Center, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
11
|
Fu W, Vasylyev D, Bi Y, Zhang M, Sun G, Khleborodova A, Huang G, Zhao L, Zhou R, Li Y, Liu S, Cai X, He W, Cui M, Zhao X, Hettinghouse A, Good J, Kim E, Strauss E, Leucht P, Schwarzkopf R, Guo EX, Samuels J, Hu W, Attur M, Waxman SG, Liu CJ. Na v1.7 as a chondrocyte regulator and therapeutic target for osteoarthritis. Nature 2024; 625:557-565. [PMID: 38172636 PMCID: PMC10794151 DOI: 10.1038/s41586-023-06888-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Osteoarthritis (OA) is the most common joint disease. Currently there are no effective methods that simultaneously prevent joint degeneration and reduce pain1. Although limited evidence suggests the existence of voltage-gated sodium channels (VGSCs) in chondrocytes2, their expression and function in chondrocytes and in OA remain essentially unknown. Here we identify Nav1.7 as an OA-associated VGSC and demonstrate that human OA chondrocytes express functional Nav1.7 channels, with a density of 0.1 to 0.15 channels per µm2 and 350 to 525 channels per cell. Serial genetic ablation of Nav1.7 in multiple mouse models demonstrates that Nav1.7 expressed in dorsal root ganglia neurons is involved in pain, whereas Nav1.7 in chondrocytes regulates OA progression. Pharmacological blockade of Nav1.7 with selective or clinically used pan-Nav channel blockers significantly ameliorates the progression of structural joint damage, and reduces OA pain behaviour. Mechanistically, Nav1.7 blockers regulate intracellular Ca2+ signalling and the chondrocyte secretome, which in turn affects chondrocyte biology and OA progression. Identification of Nav1.7 as a novel chondrocyte-expressed, OA-associated channel uncovers a dual target for the development of disease-modifying and non-opioid pain relief treatment for OA.
Collapse
Affiliation(s)
- Wenyu Fu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT, USA
| | - Yufei Bi
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Mingshuang Zhang
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Guodong Sun
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Asya Khleborodova
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Guiwu Huang
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Libo Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Renpeng Zhou
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Yonggang Li
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Shujun Liu
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT, USA
| | - Xianyi Cai
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Wenjun He
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Min Cui
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Julia Good
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Ellen Kim
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Eric Strauss
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Philipp Leucht
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Ran Schwarzkopf
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Edward X Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jonathan Samuels
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mukundan Attur
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT, USA.
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Yang J, Shin Y, Kim HJ, Kim HE, Chun JS. Prokineticin 2 is a catabolic regulator of osteoarthritic cartilage destruction in mouse. Arthritis Res Ther 2023; 25:236. [PMID: 38057865 PMCID: PMC10699050 DOI: 10.1186/s13075-023-03206-4] [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: 07/04/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Our preliminary study indicates that the multi-functional protein, prokineticin 2 (Prok2), is upregulated in osteoarthritic (OA) chondrocytes as a target of the hypoxia-inducible factor (HIF)-2α. This study aims to elucidate the potential roles of Prok2 in OA. METHODS Prok2 expression was assessed through microarray analysis in chondrocytes and confirmed via immunostaining in OA cartilage. Experimental OA was induced through destabilization of the medial meniscus (DMM). Functions of Prok2 were assessed by adenoviral overexpression, intra-articular (IA) injection of recombinant Prok2 (rProk2), and knockdown of Prok2 in joint tissues. We also explored the potential utility of Prok2 as an OA biomarker using enzyme-linked immunosorbent assay (ELISA). RESULTS HIF-2α upregulated Prok2, one of the prokineticin signaling components, in OA chondrocytes of mice and humans. Adenoviral overexpression of Prok2 in chondrocytes and cartilage explants, as well as the application of rProk2, led to an upregulation of matrix metalloproteinase (MMP)3 and MMP13. Consistently, the overexpression of Prok2 in joint tissues or IA injection of rProk2 exacerbated cartilage destruction and hindpaw mechanical allodynia induced by DMM. However, the knockdown of Prok2 in joint tissues did not significantly affect DMM-induced cartilage destruction. Additionally, despite being a secreted protein, the serum levels of Prok2 in OA mice and human OA patients were found to be below the range detected by ELISA. CONCLUSION The upregulation of Prok2 exacerbates OA cartilage destruction and hindpaw mechanical allodynia. However, its knockdown is not sufficient to inhibit experimental OA and Prok2 is not a potential candidate serum biomarker of OA.
Collapse
Affiliation(s)
- Jiye Yang
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Youngnim Shin
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hwee-Jin Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyo-Eun Kim
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jang-Soo Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
13
|
Li W, Li X, Gao Y, Xiong C, Tang Z. Emerging roles of RNA binding proteins in intervertebral disc degeneration and osteoarthritis. Orthop Surg 2023; 15:3015-3025. [PMID: 37803912 PMCID: PMC10694020 DOI: 10.1111/os.13851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/06/2023] [Accepted: 07/19/2023] [Indexed: 10/08/2023] Open
Abstract
The etiology of intervertebral disc degeneration (IDD) and osteoarthritis (OA) is complex and multifactorial. Both predisposing genes and environmental factors are involved in the pathogenesis of IDD and OA. Moreover, epigenetic modifications affect the development of IDD and OA. Dysregulated phenotypes of nucleus pulposus (NP) cells and OA chondrocytes, including apoptosis, extracellular matrix disruption, inflammation, and angiogenesis, are involved at all developmental stages of IDD and OA. RNA binding proteins (RBPs) have recently been recognized as essential post-transcriptional regulators of gene expression. RBPs are implicated in many cellular processes, such as proliferation, differentiation, and apoptosis. Recently, several RBPs have been reported to be associated with the pathogenesis of IDD and OA. This review briefly summarizes the current knowledge on the RNA-regulatory networks controlled by RBPs and their potential roles in the pathogenesis of IDD and OA. These initial findings support the idea that specific modulation of RBPs represents a promising approach for managing IDD and OA.
Collapse
Affiliation(s)
- Wen Li
- Department of EmergencyGeneral Hospital of Central Theater Command of PLAWuhanChina
| | - Xing‐Hua Li
- Department of EmergencyGeneral Hospital of Central Theater Command of PLAWuhanChina
| | - Yang Gao
- Department of OrthopaedicGeneral Hospital of Central Theater Command of PLAWuhanChina
| | - Cheng‐Jie Xiong
- Department of OrthopaedicGeneral Hospital of Central Theater Command of PLAWuhanChina
| | - Zhong‐Zhi Tang
- Department of EmergencyGeneral Hospital of Central Theater Command of PLAWuhanChina
| |
Collapse
|
14
|
Pan D, Zhong J, Zhang J, Dong H, Zhao D, Zhang H, Yao B. Function and regulation of nuclear factor 1 X-type on chondrocyte proliferation and differentiation. Gene 2023; 881:147620. [PMID: 37433356 DOI: 10.1016/j.gene.2023.147620] [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: 12/30/2022] [Revised: 05/26/2023] [Accepted: 07/05/2023] [Indexed: 07/13/2023]
Abstract
Nuclear factor 1 X-type (Nfix) is a transcription factor related to mental and physical development. However, very few studies have reported the effects of Nfix on cartilage. This study aims to reveal the influence of Nfix on the proliferation and differentiation of chondrocytes, and to explore its potential action mechanism. We isolated primary chondrocytes from the costal cartilage of newborn C57BL/6 mice and with Nfix overexpression or silencing treatment. We used Alcian blue staining and found that Nfix overexpression significantly promoted ECM synthesis in chondrocytes while silencing inhibited ECM synthesis. Using RNA-seq technology to study the expression pattern of Nfix in primary chondrocytes. We found that Nfix overexpression significantly up-regulated genes that are related to chondrocyte proliferation and extracellular matrix (ECM) synthesis and significantly down-regulated genes related to chondrocyte differentiation and ECM degradation. Nfix silencing, however, significantly up-regulated genes associated with cartilage catabolism and significantly down-regulated genes associated with cartilage growth promotion. Furthermore, Nfix exerted a positive regulatory effect on Sox9, and we propose that Nfix may promote chondrocyte proliferation and inhibit differentiation by stimulating Sox9 and its downstream genes. Our findings suggest that Nfix may be a potential target for the regulation of chondrocyte proliferation and differentiation.
Collapse
Affiliation(s)
- Daian Pan
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Jinghong Zhong
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Jingcheng Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Haisi Dong
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - He Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Baojin Yao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
15
|
Pan D, Zhong J, Zhang J, Dong H, Zhao D, Zhang H, Yao B. Function and regulation of nuclear factor 1 X-type on chondrocyte proliferation and differentiation. Gene 2023; 881:147620. [DOI: org/10.1016/j.gene.2023.147620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
|
16
|
Liu B, Wang C, Weng Z, Yang Y, Zhao H, Zhang Y, Fei Q, Shi Y, Zhang C. Glycolytic enzyme PKM2 regulates cell senescence but not inflammation in the process of osteoarthritis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1425-1433. [PMID: 37525533 PMCID: PMC10520488 DOI: 10.3724/abbs.2023062] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/19/2023] [Indexed: 04/05/2023] Open
Abstract
Chondrocyte senescence is an important mechanism underlying osteoarthritis in the senile population and is characterized by reduced expressions of the extracellular matrix proteins. The involvement of glycolysis and the tricarboxylic acid cycle in the development of osteoarthritis is inclusive. The present study aims to investigate the role of the glycolytic enzyme M2 isoform of pyruvate kinase (PKM2) in chondrocytes in senescence and inflammation. Primary chondrocytes are isolated from the knee joints of neonatal mice. Small interfering RNAs (siRNAs) against PKM2 are transfected using lipofectamine. RNA sequencing is conducted in primary chondrocytes with the PKM2 gene deleted. Cell apoptosis, autophagy, reactive oxygen species measurement, and senescent conditions are examined. The glycolytic rate in cells is measured by Seahorse examination. Interleukin 1-β (IL-1β) increases the protein expressions of matrix metallopeptidases (MMP)13 and PKM2 and reduces the protein expression of collagen type II (COL2A1) in primary chondrocytes. Silencing of PKM2 alters the protein expressions of MMP13, PKM2, and COL2A1 in the same pattern in quiescent and stimulated chondrocytes. RNA sequencing analysis reveals that PKM2 silencing reduces senescent biomarker p16 INK4a expression. Compared with low-passage chondrocytes, high-passage chondrocytes exhibit increased expression of p16 INK4a and reduced expression of COL2A1. Silencing of PKM2 reduces SA-β-Gal signals and increases COL2A1 expression in high-passage chondrocytes. Seahorse assay reveals that PKM2 deletion favors the tricarboxylic acid cycle in mitochondria in low- but not in high-passage chondrocytes. In summary, the glycolytic enzyme PMK2 modulates chondrocyte senescence but does not participate in the regulation of inflammation.
Collapse
Affiliation(s)
- Bo Liu
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Chenzhong Wang
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Ziyu Weng
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Yi Yang
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Hong Zhao
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Yueqi Zhang
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Qinming Fei
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Yi Shi
- Biomedical Research CentreZhongshan HospitalFudan UniversityShanghai200032China
| | - Chi Zhang
- Department of Orthopedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
17
|
Swahn H, Olmer M, Lotz MK. RNA-binding proteins that are highly expressed and enriched in healthy cartilage but suppressed in osteoarthritis. Front Cell Dev Biol 2023; 11:1208315. [PMID: 37457300 PMCID: PMC10349536 DOI: 10.3389/fcell.2023.1208315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Objectives: RNA-binding proteins (RBPs) have diverse and essential biological functions, but their role in cartilage health and disease is largely unknown. The objectives of this study were (i) map the global landscape of RBPs expressed and enriched in healthy cartilage and dysregulated in osteoarthritis (OA); (ii) prioritize RBPs for their potential role in cartilage and in OA pathogenesis and as therapeutic targets. Methods: Our published bulk RNA-sequencing (RNA-seq) data of healthy and OA human cartilage, and a census of 1,542 RBPs were utilized to identify RBPs that are expressed in healthy cartilage and differentially expressed (DE) in OA. Next, our comparison of healthy cartilage RNA-seq data to 37 transcriptomes in the Genotype-Tissue Expression (GTEx) database was used to determine RBPs that are enriched in cartilage. Finally, expression of RBPs was analyzed in our single cell RNA-sequencing (scRNA-seq) data from healthy and OA human cartilage. Results: Expression of RBPs was higher than nonRBPs in healthy cartilage. In OA cartilage, 188 RBPs were differentially expressed, with a greater proportion downregulated. Ribosome biogenesis was enriched in the upregulated RBPs, while splicing and transport were enriched in the downregulated. To further prioritize RBPs, we selected the top 10% expressed RBPs in healthy cartilage and those that were cartilage-enriched according to GTEx. Intersecting these criteria, we identified Tetrachlorodibenzodioxin (TCDD) Inducible Poly (ADP-Ribose) Polymerase (TIPARP) as a candidate RBP. TIPARP was downregulated in OA. scRNA-seq data revealed TIPARP was most significantly downregulated in the "pathogenic cluster". Conclusion: Our global analyses reveal expression patterns of RBPs in healthy and OA cartilage. We also identified TIPARP and other RBPs as novel mediators in OA pathogenesis and as potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Martin K. Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, United States
| |
Collapse
|
18
|
Zhang F, Dou J, Zhao X, Luo H, Ma L, Wang L, Wang Y. Identification of Key Genes Associated with Heat Stress in Rats by Weighted Gene Co-Expression Network Analysis. Animals (Basel) 2023; 13:ani13101618. [PMID: 37238049 DOI: 10.3390/ani13101618] [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: 02/20/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Heat stress has been a big challenge for animal survival and health due to global warming. However, the molecular processes driving heat stress response were unclear. In this study, we exposed the control group rats (n = 5) at 22 °C and the other three heat stress groups (five rats in each group) at 42 °C lasting 30, 60, and 120 min, separately. We performed RNA sequencing in the adrenal glands and liver and detected the levels of hormones related to heat stress in the adrenal gland, liver, and blood tissues. Weighted gene co-expression network analysis (WGCNA) was also performed. Results showed that rectal temperature and adrenal corticosterone levels were significantly negatively related to genes in the black module, which was significantly enriched in thermogenesis and RNA metabolism. The genes in the green-yellow module were strongly positively associated with rectal temperature and dopamine, norepinephrine, epinephrine, and corticosterone levels in the adrenal glands and were enriched in transcriptional regulatory activities under stress. Finally, 17 and 13 key genes in the black and green-yellow modules were identified, respectively, and shared common patterns of changes. Methyltransferase 3 (Mettl3), poly(ADP-ribose) polymerase 2 (Parp2), and zinc finger protein 36-like 1 (Zfp36l1) occupied pivotal positions in the protein-protein interaction network and were involved in a number of heat stress-related processes. Therefore, Parp2, Mettl3, and Zfp36l1 could be considered candidate genes for heat stress regulation. Our findings shed new light on the molecular processes underpinning heat stress.
Collapse
Affiliation(s)
- Fan Zhang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jinhuan Dou
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China
| | - Xiuxin Zhao
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Hanpeng Luo
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Longgang Ma
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lei Wang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yachun Wang
- College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
19
|
Xiang M, Liu L, Wu T, Wei B, Liu H. RNA-binding proteins in degenerative joint diseases: A systematic review. Ageing Res Rev 2023; 86:101870. [PMID: 36746279 DOI: 10.1016/j.arr.2023.101870] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 02/07/2023]
Abstract
RNA-binding proteins (RBPs), which are conserved proteins comprising multiple intermediate sequences, can interact with proteins, messenger RNA (mRNA) of coding genes, and non-coding RNAs to perform different biological functions, such as the regulation of mRNA stability, selective polyadenylation, and the management of non-coding microRNA (miRNA) synthesis to affect downstream targets. This article will highlight the functions of RBPs, in degenerative joint diseases (intervertebral disc degeneration [IVDD] and osteoarthritis [OA]). It will reviews the latest advancements on the regulatory mechanism of RBPs in degenerative joint diseases, in order to understand the pathophysiology, early diagnosis and treatment of OA and IVDD from a new perspective.
Collapse
Affiliation(s)
- Min Xiang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Tingrui Wu
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Bo Wei
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
20
|
Winstanley-Zarach P, Rot G, Kuba S, Smagul A, Peffers MJ, Tew SR. Analysis of RNA Polyadenylation in Healthy and Osteoarthritic Human Articular Cartilage. Int J Mol Sci 2023; 24:6611. [PMID: 37047586 PMCID: PMC10094766 DOI: 10.3390/ijms24076611] [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: 01/13/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Polyadenylation (polyA) defines the 3' boundary of a transcript's genetic information. Its position can vary and alternative polyadenylation (APA) transcripts can exist for a gene. This causes variance in 3' regulatory domains and can affect coding sequence if intronic events occur. The distribution of polyA sites on articular chondrocyte transcripts has not been studied so we aimed to define their transcriptome-wide location in age-matched healthy and osteoarthritic knee articular cartilage. Total RNA was isolated from frozen tissue samples and analysed using the QuantSeq-Reverse 3' RNA sequencing approach, where each read runs 3' to 5' from within the polyA tail into the transcript and contains a distinct polyA site. Differential expression of transcripts was significant altered between healthy and osteoarthritic samples with enrichment for functionalities that were strongly associated with joint pathology. Subsequent examination of polyA site data allowed us to define the extent of site usage across all the samples. When comparing healthy and osteoarthritic samples, we found that differential use of polyadenylation sites was modest. However, in the genes affected, there was potential for the APA to have functional relevance. We have characterised the polyadenylation landscape of human knee articular chondrocytes and conclude that osteoarthritis does not elicit a widespread change in their polyadenylation site usage. This finding differentiates knee osteoarthritis from pathologies such as cancer where APA is more commonly observed.
Collapse
Affiliation(s)
- Phaedra Winstanley-Zarach
- Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Gregor Rot
- Institute of Molecular Life Sciences, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Swiss Institute of Bioinformatics, Amphipôle, Quartier UNIL-Sorge, 1015 Lausanne, Switzerland
| | - Shweta Kuba
- Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- School of Health and Life Sciences, National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Aibek Smagul
- Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Mandy J. Peffers
- Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Simon R. Tew
- Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
21
|
Lv W, Jiang J, Xu Y, Chen Z, Wang Z, Xing A, Zheng X, Qu T, Wan Q. Re-Exploring the Inflammation-Related Core Genes and Modules in Cerebral Ischemia. Mol Neurobiol 2023; 60:3439-3451. [PMID: 36867343 DOI: 10.1007/s12035-023-03275-1] [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: 03/20/2022] [Accepted: 02/16/2023] [Indexed: 03/04/2023]
Abstract
The genetic transcription profile of brain ischemic and reperfusion injury remains elusive. To address this, we used an integrative analysis approach including differentially expressed gene (DEG) analysis, weighted-gene co-expression network analysis (WGCNA), and pathway and biological process analysis to analyze data from the microarray studies of nine mice and five rats after middle cerebral artery occlusion (MCAO) and six primary cell transcriptional datasets in the Gene Expression Omnibus (GEO). (1) We identified 58 upregulated DEGs with more than 2-fold increase, and adj. p < 0.05 in mouse datasets. Among them, Atf3, Timp1, Cd14, Lgals3, Hmox1, Ccl2, Emp1, Ch25h, Hspb1, Adamts1, Cd44, Icam1, Anxa2, Rgs1, and Vim showed significant increases in both mouse and rat datasets. (2) Ischemic treatment and reperfusion time were the main confounding factors in gene profile changes, while sampling site and ischemic time were not. (3) WGCNA identified a reperfusion-time irrelevant and inflammation-related module and a reperfusion-time relevant and thrombo-inflammation related module. Astrocytes and microglia were the main contributors of the gene changes in these two modules. (4) Forty-four module core hub genes were identified. We validated the expression of unreported stroke-associated core hubs or human stroke-associated core hubs. Zfp36 mRNA was upregulated in permanent MCAO; Rhoj, Nfkbiz, Ms4a6d, Serpina3n, Adamts-1, Lgals3, and Spp1 mRNAs were upregulated in both transient MCAO and permanent MCAO; and NFKBIZ, ZFP3636, and MAFF proteins, unreported core hubs implicated in negative regulation of inflammation, were upregulated in permanent MCAO, but not in transient MCAO. Collectively, these results expand our knowledge of the genetic profile involved in brain ischemia and reperfusion, highlighting the crucial role of inflammatory disequilibrium in brain ischemia.
Collapse
Affiliation(s)
- Wenjing Lv
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266071, China.,Department of Neurosurgery & Pathophysiology, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, 308 Ningxia Street, Qingdao, 266071, China
| | - Junqi Jiang
- Medical College, Qingdao University, Qingdao, 266071, China
| | - Yi Xu
- Medical College, Qingdao University, Qingdao, 266071, China
| | - Zhiyuan Chen
- Medical College, Qingdao University, Qingdao, 266071, China
| | - Zixuan Wang
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266071, China.,Department of Neurosurgery & Pathophysiology, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, 308 Ningxia Street, Qingdao, 266071, China
| | - Ang Xing
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266071, China
| | - Xueping Zheng
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266071, China
| | - Tingting Qu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Qi Wan
- Department of Neurosurgery & Pathophysiology, Institute of Neuroregeneration & Neurorehabilitation, Qingdao University, 308 Ningxia Street, Qingdao, 266071, China.
| |
Collapse
|
22
|
Ji ML, Li Z, Hu XY, Zhang WT, Zhang HX, Lu J. Dynamic chromatin accessibility tuning by the long noncoding RNA ELDR accelerates chondrocyte senescence and osteoarthritis. Am J Hum Genet 2023; 110:606-624. [PMID: 36868238 PMCID: PMC10119164 DOI: 10.1016/j.ajhg.2023.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/10/2023] [Indexed: 03/05/2023] Open
Abstract
Epigenetic reprogramming plays a critical role in chondrocyte senescence during osteoarthritis (OA) pathology, but the underlying molecular mechanisms remain to be elucidated. Here, using large-scale individual datasets and genetically engineered (Col2a1-CreERT2;Eldrflox/flox and Col2a1-CreERT2;ROSA26-LSL-Eldr+/+ knockin) mouse models, we show that a novel transcript of long noncoding RNA ELDR is essential for the development of chondrocyte senescence. ELDR is highly expressed in chondrocytes and cartilage tissues of OA. Mechanistically, exon 4 of ELDR physically mediates a complex consisting of hnRNPL and KAT6A to regulate histone modifications of the promoter region of IHH, thereby activating hedgehog signaling and promoting chondrocyte senescence. Therapeutically, GapmeR-mediated silencing of ELDR in the OA model substantially attenuates chondrocyte senescence and cartilage degradation. Clinically, ELDR knockdown in cartilage explants from OA-affected individuals decreased the expression of senescence markers and catabolic mediators. Taken together, these findings uncover an lncRNA-dependent epigenetic driver in chondrocyte senescence, highlighting that ELDR could be a promising therapeutic avenue for OA.
Collapse
Affiliation(s)
- Ming-Liang Ji
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, Nanjing, China.
| | - Zhuang Li
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, Nanjing, China
| | - Xin Yue Hu
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, Nanjing, China
| | - Wei Tuo Zhang
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, Nanjing, China
| | - Hai Xiang Zhang
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jun Lu
- The Center of Joint and Sports Medicine, Orthopedics Department, Zhongda Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
23
|
Li Y, Huang X, Tang J. Inhibiting the growth of ovarian cancer cells in vitro and in vivo by a small molecular inhibitor targeting La-RNA interactions. Eur J Pharmacol 2023; 940:175471. [PMID: 36549502 DOI: 10.1016/j.ejphar.2022.175471] [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: 06/13/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To identify small molecules blocking La-RNA interactions by using structural dynamics, molecular biology, and in vivo efficacy experiments. METHODS A docking virtual assay on the Chemdiv database was used to screen La binders, and their affinity were measured by surface plasmon resonance (SPR). A novel fluorescence polarization (FP) assay referring to the binding of La protein and 3'UUUOH was established to identify the inhibitors. Their activity on ovarian cancer cell proliferation, apoptosis and cell cycle were evaluated using Cell Counting Kit 8 (CCK8) and flow cytometry assay, respectively. Their in vivo efficacy against ovarian cancer growth were evaluated in a cell line-derived xenograft (CDX) model of A2780 cells. RESULTS From a total of 20 compounds with high potential binding activity with La protein, two small molecule compounds 4424-1120 and 8017-5932 with relatively stronger inhibition ability on La-RNA interactions were identified. These two compounds shared the same active centers with hydroxyimidazole and hydroxybenzene to interact with La protein through residues ARG57, GLN20 and GLN136. The in vitro assays showed that 4424-1120 and 8017-5932 effectively cause G0/G1 cell cycle arrest, inhibit cell proliferation, reduce cell invasion and promote apoptosis in ovarian cancer cells. In a CDX model on BALB/C Nude mice, we found that the growth rate of the tumor was inhibited by 4424-1120. CONCLUSION Our results demonstrated compound 4424-1120 shows good antitumor activity and safety in vitro and in vivo, and it provides a new idea for the discovery of antitumor lead compounds from small drug-like molecules.
Collapse
Affiliation(s)
- Yueyan Li
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| | - Xuan Huang
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China
| | - Jing Tang
- Department of Pharmacy, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
24
|
Gao M, Liu X, Guo P, Wang J, Li J, Wang W, Stoddart MJ, Grad S, Li Z, Wu H, Li B, He Z, Zhou G, Liu S, Zhu W, Chen D, Zou X, Zhou Z. Deciphering postnatal limb development at single-cell resolution. iScience 2023; 26:105808. [PMID: 36619982 PMCID: PMC9813795 DOI: 10.1016/j.isci.2022.105808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 08/22/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
The early postnatal limb developmental progression bridges embryonic and mature stages and mirrors the pathological remodeling of articular cartilage. However, compared with multitudinous research on embryonic limb development, the early postnatal stage seems relatively unnoticed. Here, a systematic work to portray the postnatal limb developmental landscape was carried out by characterization of 19,952 single cells from murine hindlimbs at 4 postnatal stages using single-cell RNA sequencing technique. By delineation of cell heterogeneity, the candidate progenitor sub-clusters marked by Cd34 and Ly6e were discovered in articular cartilage and enthesis, and three cellular developmental branches marked by Col10a1, Spp1, and Tnni2 were reflected in growth plate. The representative transcriptomes and developmental patterns were intensively explored, and the key regulation mechanisms as well as evolvement in osteoarthritis were discussed. Above all, these results expand horizons of postnatal limb developmental biology and reach the interconnections between limb development, remodeling, and regeneration.
Collapse
Affiliation(s)
- Manman Gao
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Department of Sport Medicine, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen 518071, China
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Peng Guo
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jianmin Wang
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Junhong Li
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Wentao Wang
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | | | - Sibylle Grad
- AO Research Institute Davos, Davos 7270, Switzerland
| | - Zhen Li
- AO Research Institute Davos, Davos 7270, Switzerland
| | - Huachuan Wu
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Baoliang Li
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhongyuan He
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Guangqian Zhou
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen 518071, China
| | - Shaoyu Liu
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Weimin Zhu
- Department of Sport Medicine, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen 518035, China
- Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Center, Shenzhen University, Shenzhen 518071, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing 100035, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhiyu Zhou
- Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
25
|
Zou L, Chu K, He X, Li Y, Zhou L, Xu X, Liao WH, Qin L. Hybrid Therapeutic Device (CUHK-OA-M2) for Relieving Symptoms Induced by Knee Osteoarthritis. Bioengineering (Basel) 2023; 10:95. [PMID: 36671667 PMCID: PMC9854676 DOI: 10.3390/bioengineering10010095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
The symptoms of knee osteoarthritis (KOA) severely affect the life quality of the elderly population. Low-level laser therapy, heat therapy, and massage therapy are widely used as independent treatments for joint disorders. However, there are very limited reports of a combination of these therapies into an integrated device for KOA so far. This study aims to develop a novel hybrid therapeutic device that can meet various requirements for knee therapy. Our hybrid therapeutic device (CUHK-OA-M2) integrated with low-level laser therapy, heat therapy, and local massage therapy can effectively provide patients with KOA with relief from their clinical symptoms. A pilot test of 50 community-dwelling elderly volunteers with KOA was performed. Finally, 43 volunteers completed two treatment periods (30 days each) and two post-treatment periods (30 days each). The Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) scores were collected and analyzed after each period. The outputs of the low-level laser, heating, and massage therapies significantly decreased the WOMAC scores in terms of pain, stiffness, function and total WOMAC after two treatment periods (p < 0.05). Although the score increased slightly after the post-treatment period, it was still lower than the baseline, indicating the treatment outcome could last for an extended period. Therefore, our CUHK-OA-M2 device, as an integrated multi-functional hybrid therapeutic device, is therapeutically significant for treating osteoarthritis symptoms on the knee joints of elderly subjects.
Collapse
Affiliation(s)
- Li Zou
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Kisum Chu
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Xuan He
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Li
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Liangbin Zhou
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiayi Xu
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, China
| | - Wei-Hsin Liao
- Department of Mechanical and Automation Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling Qin
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Liao S, Yang M, Li D, Wu Y, Sun H, Lu J, Liu X, Deng T, Wang Y, Xie N, Tang D, Nie G, Fan X. Comprehensive bulk and single-cell transcriptome profiling give useful insights into the characteristics of osteoarthritis associated synovial macrophages. Front Immunol 2023; 13:1078414. [PMID: 36685529 PMCID: PMC9849898 DOI: 10.3389/fimmu.2022.1078414] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/06/2022] [Indexed: 01/07/2023] Open
Abstract
Background Osteoarthritis (OA) is a common chronic joint disease, but the association between molecular and cellular events and the pathogenic process of OA remains unclear. Objective The study aimed to identify key molecular and cellular events in the processes of immune infiltration of the synovium in OA and to provide potential diagnostic and therapeutic targets. Methods To identify the common differential expression genes and function analysis in OA, we compared the expression between normal and OA samples and analyzed the protein-protein interaction (PPI). Additionally, immune infiltration analysis was used to explore the differences in common immune cell types, and Gene Set Variation Analysis (GSVA) analysis was applied to analyze the status of pathways between OA and normal groups. Furthermore, the optimal diagnostic biomarkers for OA were identified by least absolute shrinkage and selection operator (LASSO) models. Finally, the key role of biomarkers in OA synovitis microenvironment was discussed through single cell and Scissor analysis. Results A total of 172 DEGs (differentially expressed genes) associated with osteoarticular synovitis were identified, and these genes mainly enriched eight functional categories. In addition, immune infiltration analysis found that four immune cell types, including Macrophage, B cell memory, B cell, and Mast cell were significantly correlated with OA, and LASSO analysis showed that Macrophage were the best diagnostic biomarkers of immune infiltration in OA. Furthermore, using scRNA-seq dataset, we also analyzed the cell communication patterns of Macrophage in the OA synovial inflammatory microenvironment and found that CCL, MIF, and TNF signaling pathways were the mainly cellular communication pathways. Finally, Scissor analysis identified a population of M2-like Macrophages with high expression of CD163 and LYVE1, which has strong anti-inflammatory ability and showed that the TNF gene may play an important role in the synovial microenvironment of OA. Conclusion Overall, Macrophage is the best diagnostic marker of immune infiltration in osteoarticular synovitis, and it can communicate with other cells mainly through CCL, TNF, and MIF signaling pathways in microenvironment. In addition, TNF gene may play an important role in the development of synovitis.
Collapse
Affiliation(s)
- Shengyou Liao
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Ming Yang
- Department of Otolaryngology, Shenzhen First People’s Hospital, The Affiliated Hospital of Jinan University, Shenzhen, Guangdong, China
| | - Dandan Li
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Ye Wu
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China,Department of Otolaryngology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong Sun
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Jingxiao Lu
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Xinying Liu
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Tingting Deng
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Yujie Wang
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Ni Xie
- The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Donge Tang
- Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, the Second Clinical Medical College of Jinan University, the First Affiliated Hospital of Southern University of Science and Technology, Shenzhen People’s Hospital, Shenzhen, China
| | - Guohui Nie
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China,State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China,*Correspondence: Guohui Nie, ; Xiaoqin Fan,
| | - Xiaoqin Fan
- Shenzhen Key Laboratory of Nanozymes and Translational Cancer Research, Department of Otolaryngology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China,The Bio-bank of Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China,*Correspondence: Guohui Nie, ; Xiaoqin Fan,
| |
Collapse
|
27
|
Yin W, Lei Y, Yang X, Zou J. A two-gene random forest model to diagnose osteoarthritis based on RNA-binding protein-related genes in knee cartilage tissue. Aging (Albany NY) 2023; 15:193-212. [PMID: 36641761 PMCID: PMC9876643 DOI: 10.18632/aging.204469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/20/2022] [Indexed: 01/16/2023]
Abstract
Osteoarthritis (OA) is one of the most common diseases in the orthopedic clinic, characterized by progressive cartilage degradation. RNA-binding proteins (RBPs) are capable of binding to RNAs at transcription and translation levels, playing an important role in the pathogenesis of OA. This study aims to investigate the diagnosis values of RBP-related genes in OA. The RBPs were collected from previous studies, and the GSE114007 dataset (control = 18, OA = 20) was downloaded from the Gene Expression Omnibus (GEO) as the training cohort. Through various bioinformatical and machine learning methods, including genomic difference detection, protein-protein interaction network analyses, Lasso regression, univariate logistic regression, Boruta algorithm, and SVM-RFE, RNMT and RBM24 were identified and then included into the random forest (RF) diagnosis model. GSE117999 dataset (control = 10, OA = 10) and clinical samples collected from local hospital (control = 10, OA = 11) were used for external validation. The RF model was a promising tool to diagnose OA in the training dataset (area under curve [AUC] = 1.000, 95% confidence interval [CI] = 1.000-1.000), the GSE117999 cohort (AUC = 0.900, 95% CI = 0.769-1.000), and local samples (AUC = 0.759, 95% CI = 0.568-0.951). Besides, qPCR and Western Blotting experiments showed that RNMT (P < 0.05) and RBM24 (P < 0.01) were both down-regulated in CHON-001 cells with IL-1β treatment. In all, an RF model to diagnose OA based on RNMT and RBM24 in cartilage tissue was constructed, providing a promising clinical tool and possible cut-in points in molecular mechanism clarification.
Collapse
Affiliation(s)
- Wenhua Yin
- Department of Orthopaedics, Yuebei People’s Hospital Affiliated to Medical College of Shantou University, Shaoguan, Guangdong 512026, China
| | - Ying Lei
- Department of Audit, Yuebei People’s Hospital Affiliated to Medical College of Shantou University, Shaoguan, Guangdong 512026, China
| | - Xuan Yang
- Department of Orthopaedics, Yuebei People’s Hospital Affiliated to Medical College of Shantou University, Shaoguan, Guangdong 512026, China
| | - Jiawei Zou
- Department of Orthopaedics, Yuebei People’s Hospital Affiliated to Medical College of Shantou University, Shaoguan, Guangdong 512026, China
| |
Collapse
|
28
|
Oxymatrine Protects Chondrocytes against IL-1β-triggered Apoptosis in Vitro and Inhibits Osteoarthritis in Mice Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2745946. [PMID: 36204118 PMCID: PMC9532098 DOI: 10.1155/2022/2745946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022]
Abstract
Background Osteoarthritis (OA) is a multifactorial disease with various risk factors, resulting in the degeneration of articular cartilage and whole joints. However, to date, no effective disease-modifying therapy for OA has been developed. Oxymatrine (OMT) is associated with many pharmacological effects, including anti-inflammatory, antiapoptotic, and antioxidative properties. However, the role of OMT in OA remains unclear. Materials and Methods An IL-1β-induced chondrocyte model and anterior cruciate ligament transection (ACLT)-induced murine model of OA were constructed. The effect of OMT on chondrocyte viability was assessed using the CCK-8 assay. The protein level was assessed by Western blot analysis, and the apoptosis rate was assessed by flow cytometry in vitro and TUNEL staining in OA model mice. The effect of OMT on the degradation of articular cartilage in ACLT-induced OA mice was assessed by histological analysis. Results OMT at 0–2 mg/mL showed no conspicuous cytotoxicity on chondrocytes after 24 hours of incubation. OMT at 0.5, 1, and 2 mg/mL inhibited IL-1β-triggered apoptosis, upregulated MMP13, MMP9, and Col X, and upregulated Col II in chondrocytes in vitro. OMT represses the NF-κB signaling cascade in IL-1β-triggered chondrocytes in vitro. In an in vivo study, OMT decreased the apoptosis rate of chondrocytes and exerted a protective effect against the degradation of articular cartilage in ACLT-triggered OA mice. Conclusion OMT plays a protective role against chondrocyte injury induced by IL-1β in vitro or ACLT in vivo. OMT may play a role in chondrocytes during OA by inhibiting NF-κB signaling by decreasing the phosphorylation of p65 and IκB. OMT treatment may be a promising chondroprotective approach to delay OA cartilage progression.
Collapse
|
29
|
Chen J, Chen Z, Yuan P, Huang H, Wang J, Shi P, Sun X. ERK1 loss accelerates the progression of osteoarthritis in aged mice via NRF2/BACH1 signaling. Biochem Biophys Res Commun 2022; 622:129-135. [PMID: 35849954 DOI: 10.1016/j.bbrc.2022.07.012] [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: 06/16/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is now a common degenerative joint related disease. However, the clinical efficacy of drugs associated with cartilage regeneration remains limited. In our study, we firstly explored the role of ERK1 in the progression of OA. We clarified that ERK1-deficient mice were susceptible to age-related OA. The higher OARSI scores and more severe cartilage degeneration was observed in the ERK1-deficient mice. ERK1 deficiency decreased the nuclear transportation of Nrf2 in the chondrocytes and accelerated chondrocyte aging in vitro. Moreover, chondrocytes with ERK1 deficiency elevated the nuclear expression of BACH1, resulting in lowered expression of antioxidant enzymes in ERK1-deficient chondrocytes. The Nrf2 activator dimethyl fumarate (DMF) was used. Our experiments demonstrated the protective function of DMF against OA in ERK1 knockout mice. Above all, we confirmed the effects of ERK1 on the progression of OA and clarified the mechanisms underlying these effects. DMF might has significant use in the development of novel drugs for the therapy of OA in the future.
Collapse
Affiliation(s)
- Jian Chen
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Zhijun Chen
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Hai Huang
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiying Wang
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Peihua Shi
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| | - Xuewu Sun
- Department of Orthopaedic, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China; Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
30
|
Yi Q, Deng Z, Yue J, He J, Xiong J, Sun W, Sun W. RNA binding proteins in osteoarthritis. Front Cell Dev Biol 2022; 10:954376. [PMID: 36003144 PMCID: PMC9393224 DOI: 10.3389/fcell.2022.954376] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative joint disease worldwide. The pathological features of OA are the erosion of articular cartilage, subchondral bone sclerosis, synovitis, and metabolic disorder. Its progression is characterized by aberrant expression of genes involved in inflammation, proliferation, and metabolism of chondrocytes. Effective therapeutic strategies are limited, as mechanisms underlying OA pathophysiology remain unclear. Significant research efforts are ongoing to elucidate the complex molecular mechanisms underlying OA focused on gene transcription. However, posttranscriptional alterations also play significant function in inflammation and metabolic changes related diseases. RNA binding proteins (RBPs) have been recognized as important regulators in posttranscriptional regulation. RBPs regulate RNA subcellular localization, stability, and translational efficiency by binding to their target mRNAs, thereby controlling their protein expression. However, their role in OA is less clear. Identifying RBPs in OA is of great importance to better understand OA pathophysiology and to figure out potential targets for OA treatment. Hence, in this manuscript, we summarize the recent knowledge on the role of dysregulated RBPs in OA and hope it will provide new insight for OA study and targeted treatment.
Collapse
Affiliation(s)
- Qian Yi
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
- Department of Orthopaedics, Affiliated Hospital of Putian University, Putian, China
- Department of Physiology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jiaji Yue
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| | - Jinglong He
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| | - Jianyi Xiong
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
| | - Wei Sun
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
- *Correspondence: Wei Sun, ; Weichao Sun,
| | - Weichao Sun
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
- The Central Laboratory, Shenzhen Second People’s Hospital (The First Affiliated Hospital of Shenzhen University), Shenzhen, China
- *Correspondence: Wei Sun, ; Weichao Sun,
| |
Collapse
|
31
|
Sanchez-Lopez E, Coras R, Torres A, Lane NE, Guma M. Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol 2022; 18:258-275. [PMID: 35165404 PMCID: PMC9050956 DOI: 10.1038/s41584-022-00749-9] [Citation(s) in RCA: 291] [Impact Index Per Article: 145.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2022] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a progressive degenerative disease resulting in joint deterioration. Synovial inflammation is present in the OA joint and has been associated with radiographic and pain progression. Several OA risk factors, including ageing, obesity, trauma and mechanical loading, play a role in OA pathogenesis, likely by modifying synovial biology. In addition, other factors, such as mitochondrial dysfunction, damage-associated molecular patterns, cytokines, metabolites and crystals in the synovium, activate synovial cells and mediate synovial inflammation. An understanding of the activated pathways that are involved in OA-related synovial inflammation could form the basis for the stratification of patients and the development of novel therapeutics. This Review focuses on the biology of the OA synovium, how the cells residing in or recruited to the synovium interact with each other, how they become activated, how they contribute to OA progression and their interplay with other joint structures.
Collapse
Affiliation(s)
- Elsa Sanchez-Lopez
- Department of Orthopaedic Surgery, University of California San Diego, San Diego, CA, USA
| | - Roxana Coras
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, USA
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Alyssa Torres
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Nancy E Lane
- Division of Rheumatology, Department of Medicine, University of California Davis, Davis, CA, USA
| | - Monica Guma
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, University of California San Diego, San Diego, CA, USA.
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain.
- San Diego VA Healthcare Service, San Diego, CA, USA.
| |
Collapse
|
32
|
Chen P, Zhou J, Ruan A, Guan H, Xie J, Zeng L, Liu J, Wang Q. Synovial tissue-derived extracellular vesicles induce chondrocyte inflammation and degradation via NF-κB signalling pathway: An in vitro study. J Cell Mol Med 2022; 26:2038-2048. [PMID: 35179308 PMCID: PMC8980928 DOI: 10.1111/jcmm.17227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/09/2022] [Accepted: 01/24/2022] [Indexed: 11/28/2022] Open
Abstract
Osteoarthritis (OA) is a whole‐joint disease characterized by synovial inflammation and cartilage degeneration. However, the relationship between synovial inflammation and cartilage degeneration remains unclear. The modified Hulth's method was adopted to establish a knee OA (KOA) rabbit model. Synovial tissue was collected after 8 weeks, and synovial tissue‐derived extracellular vesicles (ST‐EVs) were extracted by filtration combined with size exclusion chromatography (SECF), followed by identification through transmission electron microscopy (TEM), nanoparticle tracer analysis (NTA) and Western blot (WB). The collagenase digestion method was used to extract normal rabbit chondrocytes, which were then treated with the SF‐EVs to observe the effect and mechanism of SF‐EVs on chondrocytes. The morphology, particle size and labelled protein marker detection confirmed that SECF successfully extract ST‐EVs. The ST‐EVs in the KOA state significantly inhibited chondrocyte proliferation and promoted chondrocytes apoptosis. Moreover, the ST‐EVs also promoted the expression of pro‐inflammatory cytokines (IL‐1β, IL‐6, TNF‐α and COX‐2) and cartilage degradation‐related enzymes (MMP13, MMP9 and ADAMTS5) in the chondrocytes. Mechanistically, the ST‐EVs significantly promoted the activation of NF‐κB signalling pathway in chondrocytes. Inhibition the activation of the NF‐κB signalling pathway significantly rescued the expression of inflammatory cytokines and cartilage degradation‐related enzymes in the ST‐EVs–induced chondrocytes. In conclusion, the ST‐EVs promote chondrocytes inflammation and degradation by activating the NF‐κB signalling pathway, providing novel insights into the occurrence and development of OA.
Collapse
Affiliation(s)
- Pu Chen
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine (The 2nd Affiliated Hospital, Guangzhou University of Chinese Medicine), Guangzhou, China.,Department of Orthopaedic Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jun Zhou
- Department of Orthopaedic Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Anmin Ruan
- Department of Orthopaedic Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China.,Department of Orthopaedic Surgery, Beijing Longfu Hospital, Beijing, China
| | - Hua Guan
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine (The 2nd Affiliated Hospital, Guangzhou University of Chinese Medicine), Guangzhou, China
| | - Jiewei Xie
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine (The 2nd Affiliated Hospital, Guangzhou University of Chinese Medicine), Guangzhou, China
| | - Lingfeng Zeng
- Department of Orthopaedic Surgery, Guangdong Provincial Hospital of Chinese Medicine (The 2nd Affiliated Hospital, Guangzhou University of Chinese Medicine), Guangzhou, China.,Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Jun Liu
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China.,Guangdong Second Traditional Chinese Medicine Hospital (Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, China
| | - Qingfu Wang
- Department of Orthopaedic Surgery, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| |
Collapse
|
33
|
Bathula CS, Chen J, Kumar R, Blackshear PJ, Saini Y, Patial S. ZFP36L1 Regulates Fgf21 mRNA Turnover and Modulates Alcoholic Hepatic Steatosis and Inflammation in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:208-225. [PMID: 34774847 PMCID: PMC8908057 DOI: 10.1016/j.ajpath.2021.10.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 10/14/2021] [Accepted: 10/29/2021] [Indexed: 02/03/2023]
Abstract
Zinc finger protein 36 like 1 (ZFP36L1) enhances the turnover of mRNAs containing AU-rich elements (AREs) in their 3'-untranslated regions (3'UTR). The physiological and pathological functions of ZFP36L1 in liver, however, remain largely unknown. Liver-specific ZFP36L1-deficient (Zfp36l1flox/flox/Cre+; L1LKO) mice were generated to investigate the role of ZFP36L1 in liver physiology and pathology. Under normal conditions, the L1LKO mice and their littermate controls (Zfp36l1flox/flox/Cre-; L1FLX) appeared normal. When fed a Lieber-DeCarli liquid diet containing alcohol, L1LKO mice were significantly protected from developing alcohol-induced hepatic steatosis, injury, and inflammation compared with L1FLX mice. Most importantly, fibroblast growth factor 21 (Fgf21) mRNA was significantly increased in the livers of alcohol diet-fed L1LKO mice compared with the alcohol diet-fed L1FLX group. The Fgf21 mRNA contains three AREs in its 3'UTR, and Fgf21 3'UTR was directly regulated by ZFP36L1 in luciferase reporter assays. Steady-state levels of Fgf21 mRNA were significantly decreased by wild-type ZFP36L1, but not by a non-binding zinc finger ZFP36L1 mutant. Finally, wild-type ZFP36L1, but not the ZFP36L1 mutant, bound to the Fgf21 3'UTR ARE RNA probe. These results demonstrate that ZFP36L1 inactivation protects against alcohol-induced hepatic steatosis and liver injury and inflammation, possibly by stabilizing Fgf21 mRNA. These findings suggest that the modulation of ZFP36L1 may be beneficial in the prevention or treatment of human alcoholic liver disease.
Collapse
Affiliation(s)
- Chandra S. Bathula
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Jian Chen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rahul Kumar
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Perry J. Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana,Address correspondence to Sonika Patial, D.V.M., Ph.D., D.A.C.V.P., Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803.
| |
Collapse
|
34
|
Yue D, Du L, Zhang B, Wu H, Yang Q, Wang M, Pan J. Time-dependently Appeared Microenvironmental Changes and Mechanism after Cartilage or Joint Damage and the Influences on Cartilage Regeneration. Organogenesis 2021; 17:85-99. [PMID: 34806543 DOI: 10.1080/15476278.2021.1991199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cartilage and joint damage easily degenerates cartilage and turns into osteoarthritis (OA), which seriously affects human life and work, and has no cure currently. The temporal and spatial changes of multiple microenvironments upon the damage of cartilage and joint are noticed, including the emergences of inflammation, bone remodeling, blood vessels, and nerves, as well as alterations of extracellular and pericellular matrix, oxygen tension, biomechanics, underneath articular cartilage tissues, and pH value. This review summarizes the existing literatures on microenvironmental changes, mechanisms, and their negative effects on cartilage regeneration following cartilage and joint damage. We conclude that time-dependently rebuilding the multiple normal microenvironments of damaged cartilage is the key for cartilage regeneration after systematic studies for the timing and correlations of various microenvironment changes.
Collapse
Affiliation(s)
- Danyang Yue
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Lin Du
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Bingbing Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Huan Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Qiong Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| | - Min Wang
- Orthopedic Department, Xinqiao Hospital, Army Medical University, Chongqing, PR China
| | - Jun Pan
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College, Chongqing University, Chongqing, PR China
| |
Collapse
|
35
|
Huang P, Wei S, Luo M, Tang Z, Lin Q, Wang X, Luo M, He Y, Wang C, Wei D, Xia C, Xu J. MiR-139-5p has an antidepressant-like effect by targeting phosphodiesterase 4D to activate the cAMP/PKA/CREB signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1594. [PMID: 34790800 PMCID: PMC8576692 DOI: 10.21037/atm-21-5149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
Background Phosphodiesterase 4D (PDE4D) inhibitor is commonly used to treat depression, but side effects seriously decrease its efficacy. PDE4D was a downstream target mRNA of miR-139-5p. Therefore, we examined the effects of hippocampal miR-139-5p gain- and loss-of-function on depression-like behaviors, the expression level of PDE4D, and hippocampus neurogenesis. Methods Bioinformatic analyses were carried out to to screen differential genes. Quantitative real-time polymerase chain reaction (qRT-PCR) and luciferase reporter assay were used to confirm the relationship between miR-139-5p and PDE4D. MiR-139-5p mimics, miR-139-5p inhibitor, or miR-NC were used to explore the function of miR-139-5p in HT-22 cells. We further explored the role of miR-139-5p in vivo using AAV-injection. Elisa, western blotting, and fluorescence in situ hybridization (FISH) were used to detect the expression of miR-139-5p and PDE4D in CRC tissues. Results Here, we showed that PDE4D messenger RNA (mRNA) was a direct target of microRNA (miR)-139-5p, which was downregulated in a chronic ultra-mild stress (CUMS)-induced depression mouse model. Moreover, in experiments in vitro, miR-139-5p mimic repressed PDE4D expression in HT-22 cells, but promoted phosphorylated cyclic-AMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) expression. Interestingly, adeno-associated virus (AAV)-miR-139-5p downregulated susceptibility to stress-induced depression-like behaviors in mice. AAV-miR-139-5p suppressed PDE4D in mouse hippocampal cells, increasing expression level of cyclic adenosine monophosphate (cAMP), p-CREB, and BDNF, and stimulating mouse hippocampal neurogenesis. Conclusions Our findings suggested that miR-139-5p acted like an antidepressant by targeting PDE4D, thereby regulating the cAMP/protein kinase A (PKA)/CREB/BDNF pathway to improve depression.
Collapse
Affiliation(s)
- Peng Huang
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Songren Wei
- Department of Neuropharmacology and Novel Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Meng Luo
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhuohong Tang
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Qingmei Lin
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Xing Wang
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Mi Luo
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Yanjun He
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Chuan Wang
- Department of Biliary Surgery, The First People's Hospital of Foshan, Foshan, China
| | - Dezhan Wei
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Chenglai Xia
- South Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China.,School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiangping Xu
- Department of Neuropharmacology and Novel Drug Discovery, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Zinc finger protein ZFP36L1 inhibits flavivirus infection by both 5'-3' XRN1 and 3'-5' RNA-exosome RNA decay pathways. J Virol 2021; 96:e0166521. [PMID: 34643435 DOI: 10.1128/jvi.01665-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Zinc-finger protein 36, CCCH type-like 1 (ZFP36L1), containing tandem CCCH-type zinc-finger motifs with an RNA-binding property, plays an important role in cellular RNA metabolism mainly via RNA decay pathways. Recently, we demonstrated that human ZFP36L1 has potent antiviral activity against influenza A virus infection. However, its role in the host defense response against flaviviruses has not been addressed. Here, we demonstrate that ZFP36L1 functions as a host innate defender against flaviviruses, including Japanese encephalitis virus (JEV) and dengue virus (DENV). Overexpression of ZFP36L1 reduced JEV and DENV infection, and ZFP36L1 knockdown enhanced viral replication. ZFP36L1 destabilized the JEV genome by targeting and degrading viral RNA mediated by both 5'-3' XRN1 and 3'-5' RNA-exosome RNA decay pathways. Mutation in both zinc-finger motifs of ZFP36L1 disrupted RNA-binding and antiviral activity. Furthermore, the viral RNA sequences specifically recognized by ZFP36L1 were mapped to the 3'-untranslated region of the JEV genome with the AU-rich element (AUUUA) motif. We extend the function of ZFP36L1 to host antiviral defense by directly binding and destabilizing the viral genome via recruiting cellular mRNA decay machineries. Importance Cellular RNA-binding proteins are among the first lines of defense against various viruses, particularly RNA viruses. ZFP36L1 belongs to the CCCH-type zinc-finger protein family and has RNA-binding activity; it has been reported to directly bind to the AU-rich elements (AREs) of a subset of cellular mRNAs and then lead to mRNA decay by recruiting mRNA degrading enzymes. However, the antiviral potential of ZFP36L1 against flaviviruses has not yet been fully demonstrated. Here, we reveal the antiviral potential of human ZFP36L1 against Japanese encephalitis virus (JEV) and dengue virus (DENV). ZFP36L1 specifically targeted the ARE motif within viral RNA and triggered the degradation of viral RNA transcripts via cellular degrading enzymes, 5'-3' XRN1 and 3'-5' RNA exosome. These findings provide mechanistic insights into how human ZFP36L1 serves as a host antiviral factor to restrict flavivirus replication.
Collapse
|
37
|
Inhibitory Effects of IL-6-Mediated Matrix Metalloproteinase-3 and -13 by Achyranthes japonica Nakai Root in Osteoarthritis and Rheumatoid Arthritis Mice Models. Pharmaceuticals (Basel) 2021; 14:ph14080776. [PMID: 34451873 PMCID: PMC8402178 DOI: 10.3390/ph14080776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 01/09/2023] Open
Abstract
Achyranthes japonica Nakai root (AJNR) is used to treat osteoarthritis (OA) and rheumatoid arthritis (RA) owing to its anti-inflammatory and antioxidant effects. This study investigated the inhibitory effects of AJNR on arthritis. AJNR was extracted using supercritical carbon dioxide (CO2), and its main compounds, pimaric and kaurenoic acid, were identified. ANJR’s inhibitory effects against arthritis were evaluated using primary cultures of articular chondrocytes and two in vivo arthritis models: destabilization of the medial meniscus (DMM) as an OA model, and collagenase-induced arthritis (CIA) as an RA model. AJNR did not affect pro-inflammatory cytokine (IL-1β, TNF-α, IL-6)-mediated cytotoxicity, but attenuated pro-inflammatory cytokine-mediated increases in catabolic factors, and recovered pro-inflammatory cytokine-mediated decreases in related anabolic factors related to in vitro. The effect of AJNR is particularly specific to IL-6-mediated catabolic or anabolic alteration. In a DMM model, AJNR decreased cartilage erosion, subchondral plate thickness, osteophyte size, and osteophyte maturity. In a CIA model, AJNR effectively inhibited cartilage degeneration and synovium inflammation in either the ankle or knee and reduced pannus formation in both the knee and ankle. Immunohistochemistry analysis revealed that AJNR mainly acted via the inhibitory effects of IL-6-mediated matrix metalloproteinase-3 and -13 in both arthritis models. Therefore, AJNR is a potential therapeutic agent for relieving arthritis symptoms.
Collapse
|
38
|
Zhou W, Shi Y, Wang H, Yu C, Zhu H, Wu A. Sinensetin Reduces Osteoarthritis Pathology in the Tert-Butyl Hydroperoxide-Treated Chondrocytes and the Destabilization of the Medial Meniscus Model Mice via the AMPK/mTOR Signaling Pathway. Front Pharmacol 2021; 12:713491. [PMID: 34335275 PMCID: PMC8322586 DOI: 10.3389/fphar.2021.713491] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/06/2021] [Indexed: 12/25/2022] Open
Abstract
As a common degenerative disease, osteoarthritis (OA) usually causes disability in the elderly and socioeconomic burden. Previous studies have shown that proper autophagy has a protective effect on OA. Sinensetin (Sin) is a methylated flavonoid derived from citrus fruits. Studies have shown that Sin is a good autophagy inducer and has shown excellent therapeutic effects in a variety of diseases; however, its role in the treatment of OA is not fully understood. This study proved the protective effect of Sin on OA through a series of in vivo and in vitro experiments. In vitro experiments have shown that Sin may inhibit chondrocyte apoptosis induced by tert-butyl hydroperoxide (TBHP); at the same time, it might also inhibit the production of MMP13 and promote the production of aggrecan and collagen II. Mechanism studies have shown that Sin promotes chondrocyte autophagy by activating AMPK/mTOR signaling pathway. On the contrary, inhibition of autophagy can partially abolish the protective effect of Sin on TBHP-treated chondrocytes. In vivo experiments show that Sin may protect against DMM-induced OA pathogenesis. These results provide evidence that Sin serves as a potential candidate for the treatment of OA.
Collapse
Affiliation(s)
- Wenxian Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Caiyu Yu
- The School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, China
| | - Huanqing Zhu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Orthopedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
39
|
Shan Q, Ma F, Wei J, Li H, Ma H, Sun P. Physiological Functions of Heat Shock Proteins. Curr Protein Pept Sci 2021; 21:751-760. [PMID: 31713482 DOI: 10.2174/1389203720666191111113726] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/24/2019] [Accepted: 09/21/2019] [Indexed: 01/03/2023]
Abstract
Heat shock proteins (HSPs) are molecular chaperones involved in a variety of life activities. HSPs function in the refolding of misfolded proteins, thereby contributing to the maintenance of cellular homeostasis. Heat shock factor (HSF) is activated in response to environmental stresses and binds to heat shock elements (HSEs), promoting HSP translation and thus the production of high levels of HSPs to prevent damage to the organism. Here, we summarize the role of molecular chaperones as anti-heat stress molecules and their involvement in immune responses and the modulation of apoptosis. In addition, we review the potential application of HSPs to cancer therapy, general medicine, and the treatment of heart disease.
Collapse
Affiliation(s)
- Qiang Shan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Fengtao Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Jingya Wei
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Hongyang Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| | - Hui Ma
- Beijing Sunlon Livestock Development Co., Ltd, Beijing, China
| | - Peng Sun
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences,
Beijing, 100193, China
| |
Collapse
|
40
|
Yang JI, Chun JS. Upregulated endonuclease Regnase-1 suppresses osteoarthritis by forming a negative feedback loop of catabolic signaling in chondrocytes. Arthritis Res Ther 2021; 23:114. [PMID: 33853646 PMCID: PMC8045248 DOI: 10.1186/s13075-021-02485-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/22/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Ribonucleases (RNases) play central roles in the post-transcriptional regulation of mRNA stability. Our preliminary results revealed that the endonuclease Regnase-1 is specifically upregulated in osteoarthritic chondrocytes. We herein explored the possible functions and regulatory mechanisms of Regnase-1 in a mouse model of osteoarthritis (OA). METHODS The expression and target genes of Regnase-1 were identified by microarray analysis in primary-culture mouse articular chondrocytes. Experimental OA in mice was induced by destabilization of the medial meniscus (DMM). The function of Regnase-1 in DMM-induced post-traumatic OA mice was examined by adenovirus-mediated overexpression or knockdown in knee joint tissues, and also by using Regnase-1 heterozygous knockout mice (Zc3h12a+/-). RESULTS Among the RNases, Regnase-1 was exclusively upregulated in chondrocytes stimulated with OA-associated catabolic factors. Adenovirus-mediated overexpression or knockdown of Regnase-1 alone in joint tissues did not cause OA-like changes. However, overexpression of Regnase-1 in joint tissues significantly ameliorated DMM-induced post-traumatic OA cartilage destruction, whereas knockdown or genetic ablation of Regnase-1 exacerbated DMM-induced cartilage destruction. Mechanistic studies suggested that Regnase-1 suppresses cartilage destruction by modulating the expression of matrix-degrading enzymes in chondrocytes. CONCLUSION Our results collectively suggest that upregulated Regnase-1 in OA chondrocytes may function as a chondro-protective effector molecule during OA pathogenesis by forming a negative feedback loop of catabolic signals, such as matrix-degrading enzyme expression, in OA chondrocytes.
Collapse
Affiliation(s)
- Jeong-In Yang
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jang-Soo Chun
- National Creative Research Initiatives Center for Osteoarthritis Pathogenesis and School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
41
|
McKellar J, Rebendenne A, Wencker M, Moncorgé O, Goujon C. Mammalian and Avian Host Cell Influenza A Restriction Factors. Viruses 2021; 13:522. [PMID: 33810083 PMCID: PMC8005160 DOI: 10.3390/v13030522] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
The threat of a new influenza pandemic is real. With past pandemics claiming millions of lives, finding new ways to combat this virus is essential. Host cells have developed a multi-modular system to detect incoming pathogens, a phenomenon called sensing. The signaling cascade triggered by sensing subsequently induces protection for themselves and their surrounding neighbors, termed interferon (IFN) response. This response induces the upregulation of hundreds of interferon-stimulated genes (ISGs), including antiviral effectors, establishing an antiviral state. As well as the antiviral proteins induced through the IFN system, cells also possess a so-called intrinsic immunity, constituted of antiviral proteins that are constitutively expressed, creating a first barrier preceding the induction of the interferon system. All these combined antiviral effectors inhibit the virus at various stages of the viral lifecycle, using a wide array of mechanisms. Here, we provide a review of mammalian and avian influenza A restriction factors, detailing their mechanism of action and in vivo relevance, when known. Understanding their mode of action might help pave the way for the development of new influenza treatments, which are absolutely required if we want to be prepared to face a new pandemic.
Collapse
Affiliation(s)
- Joe McKellar
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| | - Antoine Rebendenne
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| | - Mélanie Wencker
- Centre International de Recherche en Infectiologie, INSERM/CNRS/UCBL1/ENS de Lyon, 69007 Lyon, France;
| | - Olivier Moncorgé
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| | - Caroline Goujon
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| |
Collapse
|
42
|
Sanada Y, Tan SJO, Adachi N, Miyaki S. Pharmacological Targeting of Heme Oxygenase-1 in Osteoarthritis. Antioxidants (Basel) 2021; 10:antiox10030419. [PMID: 33803317 PMCID: PMC8001640 DOI: 10.3390/antiox10030419] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA) is a common aging-associated disease that clinically manifests as joint pain, mobility limitations, and compromised quality of life. Today, OA treatment is limited to pain management and joint arthroplasty at the later stages of disease progression. OA pathogenesis is predominantly mediated by oxidative damage to joint cartilage extracellular matrix and local cells such as chondrocytes, osteoclasts, osteoblasts, and synovial fibroblasts. Under normal conditions, cells prevent the accumulation of reactive oxygen species (ROS) under oxidatively stressful conditions through their adaptive cytoprotective mechanisms. Heme oxygenase-1 (HO-1) is an iron-dependent cytoprotective enzyme that functions as the inducible form of HO. HO-1 and its metabolites carbon monoxide and biliverdin contribute towards the maintenance of redox homeostasis. HO-1 expression is primarily regulated at the transcriptional level through transcriptional factor nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2), specificity protein 1 (Sp1), transcriptional repressor BTB-and-CNC homology 1 (Bach1), and epigenetic regulation. Several studies report that HO-1 expression can be regulated using various antioxidative factors and chemical compounds, suggesting therapeutic implications in OA pathogenesis as well as in the wider context of joint disease. Here, we review the protective role of HO-1 in OA with a focus on the regulatory mechanisms that mediate HO-1 activity.
Collapse
Affiliation(s)
- Yohei Sanada
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 7348551, Japan;
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
| | - Sho Joseph Ozaki Tan
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
| | - Nobuo Adachi
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
| | - Shigeru Miyaki
- Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 7348551, Japan;
- Department of Orthopaedic Surgery, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 7348551, Japan; (S.J.O.T.); (N.A.)
- Correspondence: ; Tel.: +81-82-257-5231
| |
Collapse
|
43
|
Zhang Y, Sun L, Liu X, Zhu D, Dang J, Xue Y, Fan H. Investigating the protective effect of tanshinone IIA against chondrocyte dedifferentiation: a combined molecular biology and network pharmacology approach. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:249. [PMID: 33708876 PMCID: PMC7940936 DOI: 10.21037/atm-20-4023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background Osteoarthritis (OA) is a common degenerative disease with multifactorial etiology. The dedifferentiation of chondrocytes can accelerate the progress of OA. Tanshinone IIA (TIIA) has been widely used to treat OA for many years and has proved to be effective in inhibiting chondrocyte dedifferentiation. Until now, the precise mechanism of TIIA’s effect against dedifferentiation has not been well understood. Methods The targets of TIIA were explored from public databases using various methods. The related targets of OA were obtained from the GeneCards database and the Online Mendelian Inheritance in Man (OMIM) database. The potential targets and signaling pathways were determined using protein-protein interaction (PPI), Gene Ontology (GO), and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Cell viability, proliferation, and metabolic activity were analyzed in vitro. The effects of TIIA on chondrocyte dedifferentiation were evaluated by assessing morphological changes, glycosaminoglycan (GAG) production, and messenger RNA (mRNA) levels of cartilage-related genes. After 48 hours of culture in medium with 100 μg/mL TIIA, chondrocytes/hydrogel spheres were implanted to repair cartilage defects in a rat model. The harvested specimens were examined with hematoxylin and eosin (H&E) staining and immunohistochemistry to evaluate cartilage regeneration. Results The results showed that there were 28 genes potentially interacting in the TIIA-chondrocyte dedifferentiation network, and nine hub genes were identified. In vitro experiments showed an inhibitory effect of TIIA on chondrocyte dedifferentiation. The proliferation and viability of chondrocytes were promoted by TIIA at a concentration of 100–200 μg/mL, but inhibited by TIIA at 400 μg/mL. Furthermore, the histology results showed that chondrocyte/hydrogel spheres pre-treated with TIIA had better cartilage repair. Conclusions This study revealed a systematic network pharmacology approach and provided a basis for the future study of TIIA as an effective treatment for cartilage regeneration. Moreover, in vitro and in vivo results confirmed the protective effects of TIIA against chondrocyte dedifferentiation.
Collapse
Affiliation(s)
- Yushen Zhang
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liguo Sun
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xincheng Liu
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Dongze Zhu
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yingsen Xue
- Department of Orthopedic Surgery, Northwest Women and Children's Hospital, Xi'an, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xi-jing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
44
|
Zhu Z, Xie J, Manandhar U, Yao X, Bian Y, Zhang B. RNA binding protein GNL3 up-regulates IL24 and PTN to promote the development of osteoarthritis. Life Sci 2021; 267:118926. [DOI: 10.1016/j.lfs.2020.118926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/26/2020] [Accepted: 12/13/2020] [Indexed: 12/17/2022]
|
45
|
Her YR, Wang L, Chepelev I, Manterola M, Berkovits B, Cui K, Zhao K, Wolgemuth DJ. Genome-wide chromatin occupancy of BRDT and gene expression analysis suggest transcriptional partners and specific epigenetic landscapes that regulate gene expression during spermatogenesis. Mol Reprod Dev 2021; 88:141-157. [PMID: 33469999 DOI: 10.1002/mrd.23449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/13/2020] [Accepted: 12/27/2020] [Indexed: 11/09/2022]
Abstract
BRDT, a member of the BET family of double bromodomain-containing proteins, is essential for spermatogenesis in the mouse and has been postulated to be a key regulator of transcription in meiotic and post-meiotic cells. To understand the function of BRDT in these processes, we first characterized the genome-wide distribution of the BRDT binding sites, in particular within gene units, by ChIP-Seq analysis of enriched fractions of pachytene spermatocytes and round spermatids. In both cell types, BRDT binding sites were mainly located in promoters, first exons, and introns of genes. BRDT binding sites in promoters overlapped with several histone modifications and histone variants associated with active transcription, and were enriched for consensus sequences for specific transcription factors, including MYB, RFX, ETS, and ELF1 in pachytene spermatocytes, and JunD, c-Jun, CRE, and RFX in round spermatids. Subsequent integration of the ChIP-seq data with available transcriptome data revealed that stage-specific gene expression programs are associated with BRDT binding to their gene promoters, with most of the BDRT-bound genes being upregulated. Gene Ontology analysis further identified unique sets of genes enriched in diverse biological processes essential for meiosis and spermiogenesis between the two cell types, suggesting distinct developmentally stage-specific functions for BRDT. Taken together, our data suggest that BRDT cooperates with different transcription factors at distinctive chromatin regions within gene units to regulate diverse downstream target genes that function in male meiosis and spermiogenesis.
Collapse
Affiliation(s)
- Yoon Ra Her
- Department of Genetics & Development, Columbia University Medical Center, New York, New York, USA
| | - Li Wang
- Department of Genetics & Development, Columbia University Medical Center, New York, New York, USA
| | - Iouri Chepelev
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.,Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Marcia Manterola
- Human Genetics Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Binyamin Berkovits
- Department of Genetics & Development, Columbia University Medical Center, New York, New York, USA
| | - Kairong Cui
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Keji Zhao
- Systems Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Debra J Wolgemuth
- Department of Genetics & Development, Columbia University Medical Center, New York, New York, USA.,Department Obstetrics & Gynecology, Columbia University Medical Center, New York, New York, USA.,Institute of Human Nutrition, Columbia University Medical Center, New York, New York, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
46
|
Chen X, Wang Y, Qu N, Zhang B, Xia C. PLCγ1 inhibition-driven autophagy of IL-1β-treated chondrocyte confers cartilage protection against osteoarthritis, involving AMPK, Erk and Akt. J Cell Mol Med 2020; 25:1531-1545. [PMID: 33372388 PMCID: PMC7875910 DOI: 10.1111/jcmm.16245] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/04/2020] [Accepted: 12/11/2020] [Indexed: 12/25/2022] Open
Abstract
Previous studies identified the involvement of phosphoinositide‐specific phospholipase C (PLC) γ1 in some events of chondrocytes. This study aims to investigate whether and how PLCγ1 modulates autophagy to execute its role in osteoarthritis (OA) progression. Rat normal or human OA chondrocytes were pretreated with IL‐1β for mimicking or sustaining OA pathological condition. Using Western blotting, immunoprecipitation, qPCR, immunofluorescence and Dimethylmethylene blue assays, and ELISA and transmission electron microscope techniques, we found that PLCγ1 inhibitor U73122 enhanced Collagen II, Aggrecan and GAG levels, accompanied with increased LC3B‐II/I ratio and decreased P62 expression level, whereas autophagy inhibitor Chloroquine partially diminished its effect. Meanwhile, U73122 dissociated Beclin1 from Beclin1‐IP3R‐Bcl‐2 complex and blocked mTOR/ULK1 axis, in which the crosstalk between PLCγ1, AMPK, Erk and Akt were involved. Additionally, by haematoxylin and eosin, Safranin O/Fast green, and immunohistochemistry staining, we observed that intra‐articular injection of Ad‐shPLCγ1‐1/2 significantly enhanced Collagen and Aggrecan levels, accompanied with increased LC3B and decreased P62 levels in a rat OA model induced by anterior cruciate ligament transection and medial meniscus resection. Consequently, PLCγ1 inhibition‐driven autophagy conferred cartilage protection against OA through promoting ECM synthesis in OA chondrocytes in vivo and in vitro, involving the crosstalk between PLCγ1, AMPK, Erk and Akt.
Collapse
Affiliation(s)
- Xiaolei Chen
- Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Yue Wang
- Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Ning Qu
- School of Medicine, Xiamen University, Xiamen, China
| | - Bing Zhang
- School of Medicine, Xiamen University, Xiamen, China
| | - Chun Xia
- Zhongshan Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
47
|
Min Y, Kim D, Suminda GGD, Zhao X, Kim M, Zhao Y, Son YO. GSK5182, 4-Hydroxytamoxifen Analog, a New Potential Therapeutic Drug for Osteoarthritis. Pharmaceuticals (Basel) 2020; 13:ph13120429. [PMID: 33261216 PMCID: PMC7761342 DOI: 10.3390/ph13120429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 11/16/2022] Open
Abstract
Estrogen-related receptors (ERRs) are the first identified orphan nuclear receptors. The ERR family consists of ERRα, ERRβ, and ERRγ, regulating diverse isoform-specific functions. We have reported the importance of ERRγ in osteoarthritis (OA) pathogenesis. However, therapeutic approaches with ERRγ against OA associated with inflammatory mechanisms remain limited. Herein, we examined the therapeutic potential of a small-molecule ERRγ inverse agonist, GSK5182 (4-hydroxytamoxifen analog), in OA, to assess the relationship between ERRγ expression and pro-inflammatory cytokines in mouse articular chondrocyte cultures. ERRγ expression increased following chondrocyte exposure to various pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. Pro-inflammatory cytokines dose-dependently increased ERRγ protein levels. In mouse articular chondrocytes, adenovirus-mediated ERRγ overexpression upregulated matrix metalloproteinase (MMP)-3 and MMP-13, which participate in cartilage destruction during OA. Adenovirus-mediated ERRγ overexpression in mouse knee joints or ERRγ transgenic mice resulted in OA. In mouse joint tissues, genetic ablation of Esrrg obscured experimental OA. These results indicate that ERRγ is involved in OA pathogenesis. In mouse articular chondrocytes, GSK5182 inhibited pro-inflammatory cytokine-induced catabolic factors. Consistent with the in vitro results, GSK5182 significantly reduced cartilage degeneration in ERRγ-overexpressing mice administered intra-articular Ad-Esrrg. Overall, the ERRγ inverse agonist GSK5182 represents a promising therapeutic small molecule for OA.
Collapse
Affiliation(s)
- Yunhui Min
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Dahye Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
| | - Godagama Gamaarachchige Dinesh Suminda
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Xiangyu Zhao
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
| | - Mangeun Kim
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
| | - Yaping Zhao
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju City 63243, Korea; (Y.M.); (G.G.D.S.); (X.Z.)
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju City 63243, Korea; (D.K.); (M.K.)
- Bio-Health Materials Core-Facility Center, Jeju National University, Jeju City 63243, Korea
- Practical Translational Research Center, Jeju National University, Jeju City 63243, Korea
- Correspondence: ; Tel.: +82-(64)-754-3331
| |
Collapse
|
48
|
Feng X, Zhou S, Cai W, Guo J. The miR-93-3p/ZFP36L1/ZFX axis regulates keratinocyte proliferation and migration during skin wound healing. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:450-463. [PMID: 33473330 PMCID: PMC7803633 DOI: 10.1016/j.omtn.2020.11.017] [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: 06/14/2020] [Accepted: 11/17/2020] [Indexed: 01/01/2023]
Abstract
Keratinocyte proliferation and migration are crucial steps during skin wound healing. The functional role of microRNAs (miRs) remains relatively unknown during this process. miR-93 levels have been reported to increase within 24 h of skin wound healing; however, whether miR-93-3p or miR-93-5p plays a specific role in wound healing is yet to be studied. In this study, with the use of an in vivo mouse skin wound-healing model, we demonstrate that miR-93-3p is significantly upregulated, whereas there is no change in the expression of miR-93-5p during skin wound healing. In HaCaT cells, miR-93-3p overexpression increased proliferation and migration of the cells, whereas miR-93-3p inhibition had the reverse effect. Additionally, it was evident that ZFP36L1 was a direct target of miR-93-3p in keratinocytes. Further, ZFP36L1 silencing mirrored the consequences observed during miR-93-3p overexpression on both proliferation and migration of keratinocytes. In addition, we demonstrate that zinc-finger X-linked (ZFX), as a target for ZFP36L1, is involved in the promotion of the miR-93-3p/ZFP36L1 axis in keratinocyte proliferation and migration. Ultimately, we found that mouse skin wound model treatment with anti-miR-93-3p delayed wound healing. Overall, our results show that miR-93-3p is a crucial regulator of skin wound healing that facilitates keratinocyte proliferation and migration through ZFP36L1/ZFX axis.
Collapse
Affiliation(s)
- Xiao Feng
- Department of Plastic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Shuangbai Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Weilin Cai
- Department of Dermatologic Surgery, Hangzhou Third People's Hospital, Hangzhou, China
| | - Jincai Guo
- Department of Plastic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| |
Collapse
|
49
|
Guo J, Lei M, Cheng F, Liu Y, Zhou M, Zheng W, Zhou Y, Gong R, Liu Z. RNA-binding proteins tristetraprolin and human antigen R are novel modulators of podocyte injury in diabetic kidney disease. Cell Death Dis 2020; 11:413. [PMID: 32487989 PMCID: PMC7265504 DOI: 10.1038/s41419-020-2630-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022]
Abstract
Diabetic kidney disease (DKD) is one of the most common complications of diabetes, and the most common cause of end-stage renal disease, for which no effective therapies are yet available. RNA-binding proteins (RBPs) play a pivotal role in epigenetic regulation; tristetraprolin (TTP) and human antigen R (HuR) competitively bind cytokine mRNAs, exert contrasting effects on RNA stability, and drive inflammation. However, RBPs’ roles in diabetes-related glomerulopathy are poorly understood. Herein, we investigated whether TTP and HuR are involved in post-transcriptional regulation of podocytopathic molecules and inflammatory cytokines in DKD. In DKD patients and db/db mice, TTP expression was significantly decreased and HuR expression was increased in glomerular podocytes, concurrent with podocyte injury, histological signs of DKD, and augmented glomerular expression of interleukin (IL)-17 and claudin-1, which are targets of TTP and HuR, as evidenced by RNA immunoprecipitation. In cultured podocytes, exposure to high ambient glucose amplified HuR expression and repressed TTP expression, upregulated IL-17 and claudin-1, and promoted podocyte injury. Thus, TTP hypoactivity or HuR hyperactivity is sufficient and essential to diabetic podocytopathy. Moreover, in silico analysis revealed that several kinases govern phosphorylation and activation of TTP and HuR, and glycogen synthase kinase (GSK)-3β activated both TTP and HuR, which harbor putative GSK-3β consensus phosphorylation motifs. Treatment of db/db mice with a small molecule inhibitor of GSK-3β abrogated the changes in TTP and HuR in glomeruli and mitigated the overexpression of their target genes (IL-17, claudin-1, B7-1, and MCP-1) thus also mitigating proteinuria and DKD pathology. Our study indicates that TTP and HuR are dysregulated in DKD via a GSK-3β-mediated mechanism and play crucial roles in podocyte injury through post-transcriptional regulation of diverse genes. It also provides novel insights into DKD’s pathophysiology and identifies potential therapeutic targets.
Collapse
Affiliation(s)
- Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.,Division of Kidney disease and Hypertension, Brown Medical School, Providence, RI, 02903, USA.,Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, USA
| | - Min Lei
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Fei Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yong Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mengwen Zhou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Wen Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yali Zhou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Rujun Gong
- Division of Kidney disease and Hypertension, Brown Medical School, Providence, RI, 02903, USA. .,Division of Nephrology, Department of Medicine, University of Toledo College of Medicine, Toledo, OH, 43614, USA.
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
50
|
Rajapakse D, Peterson K, Mishra S, Fan J, Lerner J, Campos M, Wistow G. Amelotin is expressed in retinal pigment epithelium and localizes to hydroxyapatite deposits in dry age-related macular degeneration. Transl Res 2020; 219:45-62. [PMID: 32160961 PMCID: PMC7197213 DOI: 10.1016/j.trsl.2020.02.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 11/28/2022]
Abstract
Deposition of hydroxyapatite (HAP) basal to the retinal pigment epithelium (RPE) is linked to the progression of age-related macular degeneration (AMD). Serum-deprivation of RPE cells in culture mimics some features of AMD. We now show that serum-deprivation also leads to the induction of amelotin (AMTN), a protein involved in hydroxyapatite mineralization in enamel. HAP is formed in our culture model and is blocked by siRNA inhibition of AMTN expression. In situ hybridization and immunofluorescence imaging of human eye tissue show that AMTN is expressed in RPE of donor eyes with geographic atrophy ("dry" AMD) in regions with soft drusen containing HAP spherules or nodules. AMTN is not found in hard drusen, normal RPE, or donor eyes diagnosed with wet AMD. These findings suggest that AMTN is involved in formation of HAP spherules or nodules in AMD, and as such provides a new therapeutic target for slowing disease progression.
Collapse
Affiliation(s)
- Dinusha Rajapakse
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Katherine Peterson
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Sanghamitra Mishra
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Jianguo Fan
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Joshua Lerner
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Campos
- Histopathology Core Facility, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Graeme Wistow
- Section on Molecular Structure and Functional Genomics, National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|