1
|
Ziaastani Z, Kalantari-Khandani B, Niazi MJ, Kazemipour A. Identification of critical genes and metabolic pathways in rheumatoid arthritis and osteoporosis toward drug repurposing. Comput Biol Med 2024; 180:108912. [PMID: 39079412 DOI: 10.1016/j.compbiomed.2024.108912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) and osteoporosis (OP) are considered to be complex diseases. In recent studies, a positive association between RA and OP has been reported triggering growing research interest. This study aims to investigate the drugs related to critical genes in RA and OP, using bioinformatics approaches, toward drug repurposing. METHOD RA and OP genes were identified. The RA-OP PPI network was constructed and analyzed using the STRING and Cytoscape, respectively. Hub genes and modules were extracted and enriched Gene Ontology, through the WebGestalt and g:Profiler. The identification of the drugs related to critical genes using the DGIDB, and extracted the miRNAs using miRWalk and miRNet. RESULTS By network clustering, five significant modules were obtained that have important roles in the immune system. IL6, TNF, IL1B, STAT3, TGFB1, TP53, HIF1A, CCL2, IL10, and MMP9 were found as the top 10 hub genes in the RA-OP network. Hub genes were shown to have implications in inflammatory response, significant functions in cytokine receptor binding, and localized mostly in extracellular space. By investigating the drugs related to hub genes, 16 drugs were identified as repurposing candidate drugs. The 10 drugs included Hydroxychloroquine, Infliximab, Adalimumab, Etanercept, Certolizumab, Cyclosporine, Diacerein, Gevokizumab, Canakinumab, and Olokizumab proposed for OP. Also, six drugs including Pirfenidone, Pentoxifylline, Vadimezan, Rilonacept, Metelimumab, and Siltuximab have important roles in inflammatory control and were proposed for both RA and OP. CONCLUSIONS The results of the present study can provide novel insights into the pathogenesis and treatment of RA and OP.
Collapse
Affiliation(s)
- Zahra Ziaastani
- Faculty of Mathematics and Computer Science, Department of Bioinformatics, Bahonar University, Kerman, Iran; Bahonar Bioinformatics Lab (BBL), Iran
| | | | - Mohammad-Javad Niazi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran; Bahonar Bioinformatics Lab (BBL), Iran
| | - Ali Kazemipour
- Faculty of Mathematics and Computer Science, Department of Bioinformatics, Bahonar University, Kerman, Iran; Bahonar Bioinformatics Lab (BBL), Iran.
| |
Collapse
|
2
|
Chen L, Wu T, Zhang M, Ding Z, Zhang Y, Yang Y, Zheng J, Zhang X. [Identification of potential biomarkers and immunoregulatory mechanisms of rheumatoid arthritis based on multichip co-analysis of GEO database]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1098-1108. [PMID: 38977339 PMCID: PMC11237296 DOI: 10.12122/j.issn.1673-4254.2024.06.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
OBJECTIVE To identify the biomarkers for early rheumatoid arthritis (RA) diagnosis and explore the possible immune regulatory mechanisms. METHODS The differentially expressed genesin RA were screened and functionally annotated using the limma, RRA, batch correction, and clusterProfiler. The protein-protein interaction network was retrieved from the STRING database, and Cytoscape 3.8.0 and GeneMANIA were used to select the key genes and predicting their interaction mechanisms. ROC curves was used to validate the accuracy of diagnostic models based on the key genes. The disease-specific immune cells were selected via machine learning, and their correlation with the key genes were analyzed using Corrplot package. Biological functions of the key genes were explored using GSEA method. The expression of STAT1 was investigated in the synovial tissue of rats with collagen-induced arthritis (CIA). RESULTS We identified 9 core key genes in RA (CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL), which regulate synovial inflammation primarily through cytokines-related pathways. ROC curve analysis showed a high predictive accuracy of the 9 core genes, among which STAT1 had the highest AUC (0.909). Correlation analysis revealed strong correlations of CD3G, ITGAL, LCK, CD8A, and STAT1 with disease-specific immune cells, and STAT1 showed the strongest correlation with M1-type macrophages (R=0.68, P=2.9e-08). The synovial tissues of the ankle joints of CIA rats showed high expressions of STAT1 and p-STAT1 with significant differential expression of STAT1 between the nucleus and the cytoplasm of the synovial fibroblasts. The protein expressions of p-STAT1 and STAT1 in the cell nuclei were significantly reduced after treatment. CONCLUSION CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL may serve as biomarkers for early diagnosis of RA. Gene-immune cell pathways such as CD3G/CD8A/LCK-γδ T cells, ITGAL-Tfh cells, and STAT1-M1-type macrophages may be closely related with the development of RA.
Collapse
Affiliation(s)
- L Chen
- School of Health Management, Bengbu Medical University, Bengbu 233030, China
| | - T Wu
- School of Public Health, Bengbu Medical University, Bengbu 233030, China
| | - M Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu 233030, China
- School of Sports Medicine and Rehabilitation, Shandong First Medical University, Taian 271016, China
| | - Z Ding
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233030, China
| | - Y Zhang
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233030, China
| | - Y Yang
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233030, China
| | - J Zheng
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233030, China
| | - X Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu Medical University, Bengbu 233030, China
| |
Collapse
|
3
|
Ramírez-Solano MA, Córdova EJ, Orozco L, Tejero ME. Plasma MicroRNAs Related to Metabolic Syndrome in Mexican Women. Lifestyle Genom 2023; 16:165-176. [PMID: 37708875 DOI: 10.1159/000534041] [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: 07/15/2022] [Accepted: 09/01/2023] [Indexed: 09/16/2023] Open
Abstract
INTRODUCTION The metabolic syndrome (MetS) is a cluster of abnormalities related to cardiovascular disease (CVD). Circulating miRNAs (c-miRNAs) are non-coding RNAs associated with different phenotypes, some of them integrating the MetS. The aim of the study was to compare the c-miRNAs profile in plasma between women with MetS and controls and explore their possible association with dysregulation of metabolic pathways. METHODS The study was conducted in two phases. At the screening phase, miRNA composition in fasting plasma was compared between 8 participants with MetS and 10 healthy controls, using microarray technology. The validation phase included the analysis by qRT-PCR of 10 selected c-miRNAs in an independent sample (n = 29). RESULTS We found 21 c-miRNAs differentially expressed between cases and controls. The concentration in plasma of the c-miRNAs hsa-miR-1260a, hsa-miR-4514, and hsa-miR-4687-5p were also correlated with risk factors for CVD. Differences of hsa-miR-1260a between cases and controls were validated using qRT-PCR (fold-change = 7.0; p = 0.003). CONCLUSION The signature of plasma c-miRNAs differed between women with MetS and controls. The identified miRNAs regulate pathways related to the MetS such as insulin resistance and adipokine activity. The role of c-miR-1260a in the MetS remains to be elucidated.
Collapse
Affiliation(s)
- Marisol Adelina Ramírez-Solano
- Laboratorio de Nutrigenómica y Nutrigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
- Maestría en Bioquímica Clínica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Emilio J Córdova
- Consorcio Oncogenómica y Enfermedades Óseas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Lorena Orozco
- Laboratorio de Inmunogenómica y Enfermedades Complejas, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - María Elizabeth Tejero
- Laboratorio de Nutrigenómica y Nutrigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
4
|
Gao Y, Cai W, Zhou Y, Li Y, Cheng J, Wei F. Immunosenescence of T cells: a key player in rheumatoid arthritis. Inflamm Res 2022; 71:1449-1462. [DOI: 10.1007/s00011-022-01649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/05/2022] Open
|
5
|
Li W, Wang K, Liu Y, Wu H, He Y, Li C, Wang Q, Su X, Yan S, Su W, Zhang Y, Lin N. A Novel Drug Combination of Mangiferin and Cinnamic Acid Alleviates Rheumatoid Arthritis by Inhibiting TLR4/NFκB/NLRP3 Activation-Induced Pyroptosis. Front Immunol 2022; 13:912933. [PMID: 35799788 PMCID: PMC9253268 DOI: 10.3389/fimmu.2022.912933] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022] Open
Abstract
Growing evidence shows that Baihu-Guizhi decoction (BHGZD), a traditional Chinese medicine (TCM)-originated disease-modifying anti-rheumatic prescription, may exert a satisfying clinical efficacy for rheumatoid arthritis (RA) therapy. In our previous studies, we verified its immunomodulatory and anti-inflammatory activities. However, bioactive compounds (BACs) of BHGZD and the underlying mechanisms remain unclear. Herein, an integrative research strategy combining UFLC-Q-TOF-MS/MS, gene expression profiling, network calculation, pharmacokinetic profiling, surface plasmon resonance, microscale thermophoresis, and pharmacological experiments was carried out to identify the putative targets of BHGZD and underlying BACs. After that, both in vitro and in vivo experiments were performed to determine the drug effects and pharmacological mechanisms. As a result, the calculation and functional modularization based on the interaction network of the “RA-related gene–BHGZD effective gene” screened the TLR4/PI3K/AKT/NFκB/NLRP3 signaling-mediated pyroptosis to be one of the candidate effective targets of BHGZD for reversing the imbalance network of “immune-inflammation” during RA progression. In addition, both mangiferin (MG) and cinnamic acid (CA) were identified as representative BACs acting on that target, for the strong binding affinities between compounds and target proteins, good pharmacokinetic features, and similar pharmacological effects to BHGZD. Notably, both BHGZD and the two-BAC combination of MG and CA effectively alleviated the disease severity of the adjuvant-induced arthritis-modified rat model, including elevating pain thresholds, relieving joint inflammation and bone erosion via inhibiting NF-κB via TLR4/PI3K/AKT signaling to suppress the activation of the NLRP3 inflammasome, leading to the downregulation of downstream caspase-1, the reduced release of IL-1β and IL-18, and the modulation of GSDMD-mediated pyroptosis. Consistent data were obtained based on the in vitro pyroptosis cellular models of RAW264.7 and MH7A cells induced by LPS/ATP. In conclusion, these findings offer an evidence that the MG and CA combination identified from BHGZD may interact with TLR4/PI3K/AKT/NFκB signaling to inhibit NLRP3 inflammasome activation and modulate pyroptosis, which provides the novel representative BACs and pharmacological mechanisms of BHGZD against active RA. Our data may shed new light on the mechanisms of the TCM formulas and promote the modernization development of TCM and drug discovery.
Collapse
Affiliation(s)
- Weijie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kexin Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yudong Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Wu
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yan He
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Congchong Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaohui Su
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shikai Yan
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Weiwei Su
- Guangdong Engineering and Technology Research Center for Quality and Efficacy Reevaluation of Post-Market Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanqiong Zhang, ; Na Lin,
| | - Na Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yanqiong Zhang, ; Na Lin,
| |
Collapse
|
6
|
Kmiołek T, Paradowska-Gorycka A. miRNAs as Biomarkers and Possible Therapeutic Strategies in Rheumatoid Arthritis. Cells 2022; 11:cells11030452. [PMID: 35159262 PMCID: PMC8834522 DOI: 10.3390/cells11030452] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023] Open
Abstract
Within the past years, more and more attention has been devoted to the epigenetic dysregulation that provides an additional window for understanding the possible mechanisms involved in the pathogenesis of autoimmune rheumatic diseases. Rheumatoid arthritis (RA) is a heterogeneous disease where a specific immunologic and genetic/epigenetic background is responsible for disease manifestations and course. In this field, microRNAs (miRNA; miR) are being identified as key regulators of immune cell development and function. The identification of disease-associated miRNAs will introduce us to the post-genomic era, providing the real probability of manipulating the genetic impact of autoimmune diseases. Thereby, different miRNAs may be good candidates for biomarkers in disease diagnosis, prognosis, treatment and other clinical applications. Here, we outline not only the role of miRNAs in immune and inflammatory responses in RA, but also present miRNAs as diagnostic/prognostic biomarkers. Research into miRNAs is still in its infancy; however, investigation into these novel biomarkers could progress the use of personalized medicine in RA treatment. Finally, we discussed the possibility of miRNA-based therapy in RA patients, which holds promise, given major advances in the therapy of patients with inflammatory arthritis.
Collapse
|
7
|
Caputo V, Strafella C, Termine A, Fabrizio C, Ruffo P, Cusumano A, Giardina E, Ricci F, Cascella R. Epigenomic signatures in age-related macular degeneration: Focus on their role as disease modifiers and therapeutic targets. Eur J Ophthalmol 2021; 31:2856-2867. [PMID: 34798695 DOI: 10.1177/11206721211028054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Epigenetics is characterized by molecular modifications able to shape gene expression profiles in response to inner and external stimuli. Therefore, epigenetic elements are able to provide intriguing and useful information for the comprehension and management of different human conditions, including aging process, and diseases. On this subject, Age-related Macular Degeneration (AMD) represents one of the most frequent age-related disorders, dramatically affecting the quality of life of older adults worldwide. The etiopathogenesis is characterized by an interplay among multiple genetic and non-genetic factors, which have been extensively studied. Nevertheless, a deeper dissection of molecular machinery associated with risk, onset, progression and effectiveness of therapies is still missing. In this regard, epigenetic signals may be further explored to disentangle disease etiopathogenesis, the possible therapeutic avenues and the differential response to AMD treatment. This review will discuss the epigenomic signatures mostly investigated in AMD, which could be applied to improve the knowledge of disease mechanisms and to set-up novel or modified treatment options.
Collapse
Affiliation(s)
- Valerio Caputo
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy.,Medical Genetics Laboratory, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Claudia Strafella
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy.,Medical Genetics Laboratory, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Andrea Termine
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Carlo Fabrizio
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Paola Ruffo
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Andrea Cusumano
- UOSD of Ophthalmology PTV Foundation "Policlinico Tor Vergata", Rome, Italy
| | - Emiliano Giardina
- Genomic Medicine Laboratory UILDM, IRCCS Santa Lucia Foundation, Rome, Italy.,UILDM Lazio ONLUS Foundation, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Federico Ricci
- UNIT Retinal Diseases PTV Foundation "Policlinico Tor Vergata", Rome, Italy
| | - Raffaella Cascella
- Medical Genetics Laboratory, Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy.,Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, Tirana, Albania
| |
Collapse
|
8
|
Ali SA, Peffers MJ, Ormseth MJ, Jurisica I, Kapoor M. The non-coding RNA interactome in joint health and disease. Nat Rev Rheumatol 2021; 17:692-705. [PMID: 34588660 DOI: 10.1038/s41584-021-00687-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 02/07/2023]
Abstract
Non-coding RNAs have distinct regulatory roles in the pathogenesis of joint diseases including osteoarthritis (OA) and rheumatoid arthritis (RA). As the amount of high-throughput profiling studies and mechanistic investigations of microRNAs, long non-coding RNAs and circular RNAs in joint tissues and biofluids has increased, data have emerged that suggest complex interactions among non-coding RNAs that are often overlooked as critical regulators of gene expression. Identifying these non-coding RNAs and their interactions is useful for understanding both joint health and disease. Non-coding RNAs regulate signalling pathways and biological processes that are important for normal joint development but, when dysregulated, can contribute to disease. The specific expression profiles of non-coding RNAs in various disease states support their roles as promising candidate biomarkers, mediators of pathogenic mechanisms and potential therapeutic targets. This Review synthesizes literature published in the past 2 years on the role of non-coding RNAs in OA and RA with a focus on inflammation, cell death, cell proliferation and extracellular matrix dysregulation. Research to date makes it apparent that 'non-coding' does not mean 'non-essential' and that non-coding RNAs are important parts of a complex interactome that underlies OA and RA.
Collapse
Affiliation(s)
- Shabana A Ali
- Bone and Joint Center, Department of Orthopaedic Surgery, Henry Ford Health System, Detroit, MI, USA. .,Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, USA.
| | - Mandy J Peffers
- Department of Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Michelle J Ormseth
- Department of Research and Development, Veterans Affairs Medical Center, Nashville, TN, USA.,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Igor Jurisica
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Data Science Discovery Centre for Chronic Diseases, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
9
|
Zhuo Q, Wei L, Yin X, Li H, Qin G, Li S, Peng TT, Liu B, Zhao S, Ye Z. LncRNA ZNF667-AS1 alleviates rheumatoid arthritis by sponging miR-523-3p and inactivating the JAK/STAT signalling pathway. Autoimmunity 2021; 54:406-414. [PMID: 34423698 DOI: 10.1080/08916934.2021.1966770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease, which compromises the synovial membrane resulting in chronic inflammation. Increasing evidence has demonstrated that long non-coding RNAs (lncRNAs) are implicated in the pathogenesis of RA. This study investigated the role of lncRNA ZNF667-AS1 in RA progression. METHODS Synovial tissues and fibroblast-like synoviocytes (FLSs) were obtained from patients with RA. Gene expression was measured using RT-qPCR. Chondrocytes were treated with lipopolysaccharide (LPS) to establish in vitro models of OA. Cell counting kit-8 (CCK-8), western blot, and enzyme-linked immunosorbent assay (ELISA) were used to examine the proliferation and inflammatory cytokine production in chondrocytes. Animal models of OA were established in SD rats. Peripheral blood mononuclear cells (PBMCs) were isolated from the OA rats. Flow cytometry was used to measure the changes of the inflammatory T-helper cell 17 (Th17) cells. The relationship between ZNF667-AS1 and miR-523-3p was verified by luciferase reporter assay. RESULTS ZNF667-AS1 was downregulated in RA-FLSs and LPS-stimulated chondrocytes. ZNF667-AS1 overexpression significantly promoted cell proliferation and inhibited the production of IL-6, IL-17 and TNF-α in LPS-stimulated chondrocytes. Additionally, ZNF667-AS1 overexpression reduced the generation of CD4 + IL-17+ cells. In mechanism, ZNF667-AS1 acted a sponge for miR-523-3p. MiR-523-3p overexpression reversed the ZNF667-AS1-mediated regulation of cell proliferation and inflammation. Furthermore, miR-523-3p overexpression abolished the inhibitory effects of ZNF667-AS1 on the JAK/STAT signalling activation. CONCLUSION ZNF667-AS1 exerts protective effects during RA development by sponging miR-523-3p and inactivating the JAK/STAT signalling.
Collapse
Affiliation(s)
- Qin Zhuo
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China.,Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Lu Wei
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Xietian Yin
- The First Clinical College, Hubei University of Chinese Medicine, Wuhan, China.,Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Huiling Li
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Guifu Qin
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Siqi Li
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Ting Ting Peng
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Bo Liu
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Shichao Zhao
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Zhiqin Ye
- Department of Rheumatism Immunology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
10
|
Wang Y, Yin Z, Zhang N, Song H, Zhang Q, Hao X, Wang Z. MiR-125a-3p inhibits cell proliferation and inflammation responses in fibroblast-like synovial cells in rheumatoid arthritis by mediating the Wnt/β-catenin and NF-κB pathways via targeting MAST3. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2021; 21:560-567. [PMID: 34854396 PMCID: PMC8672414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVES To explore the role and mechanism of miR-125a-3p in rheumatoid arthritis (RA) progression. METHODS The RA-tissues and fibroblast-like synovial cells in rheumatoid arthritis (RA-FLS) were used in this study. qRT-PCR, western blot and ELISA assay were performed to detect the expression levels of IL-6, IL-β and ΤΝF-α. Dual-luciferase reporter gene assay was used to observe the binding effect of miR-125a-3p and MAST3, and CCK-8 was used to observe the effect of miR-125a-3p on the proliferation of RA-FLS. RESULTS miR-125a-3p was significantly downregulated in the RA-tissues and RA-FLS, and miR-125a-3p could inhibit the proliferation and reduce the inflammation response of RA-FLS. Besides, MAST3 was found as a target of miR-125a-3p, and increased MAST3 could reverse the effects of miR-125a-3p on RA-FLS including decreased proliferation, reduced inflammation level and the inactivation of Wnt/β-catenin and NF-κB pathways. CONCLUSIONS This study suggests that miR-125a-3p could inactivate the Wnt/β-catenin and NF-κB pathways to reduce the proliferation and inflammation response of RA-FLS via targeting MAST3.
Collapse
Affiliation(s)
- Yingxue Wang
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China
| | - Zhe Yin
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China
| | - Ni Zhang
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China
| | - Huishu Song
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China
| | - Qiuting Zhang
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China
| | - Xuexi Hao
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China
| | - Zhilun Wang
- Department of Rheumatology, The Second Hospital of Shandong University, Shandong Province, China,Corresponding author: Zhilun Wang, Department of Rheumatology, The Second Hospital of Shandong University, No. 247 Beiyuan Street, Tianqiao District, Jinan 250033, Shandong Province, China E-mail:
| |
Collapse
|