1
|
Zhang MY, Zhou MY, Gong XN, Zhou YX, Li LL, Xu ZS, Feng HY, Lv M, Li R, Cai L. Acetazolamide as an aquaporin 1 inhibitor mitigates rheumatoid arthritis by reducing angiogenesis via the modulation of the FAK-PI3K/Akt signaling pathway. Int Immunopharmacol 2025; 158:114845. [PMID: 40359885 DOI: 10.1016/j.intimp.2025.114845] [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: 12/03/2024] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Synovial angiogenesis worsens the course of rheumatoid arthritis (RA) by driving pannus formation and chronic inflammation, implying anti-angiogenesis as a possible treatment strategy. Aquaporin 1 (AQP1) is a promising target for RA, and its inhibitor acetazolamide (AZ) exerts anti-arthritic effects in RA animal models. Particularly, AZ is effective in inhibiting angiogenesis in multiple illnesses. However, AZ's impacts on angiogenesis in the RA context and its underlying mechanisms are unclear. This study investigated AZ's influences on migration, invasion, chemotaxis, and tube formation in human umbilical vein endothelial cells (HUVECs). Moreover, AZ's anti-angiogenic effects were examined in zebrafish and adjuvant-induced arthritis (AIA) rat synovium. The involvement of the focal adhesion kinase (FAK)-PI3K/AKT axis was also studied. Herein, AZ inhibited VEGF-induced migration and invasion in HUVECs and reduced their chemotactic migration toward RA fibroblast-like synoviocytes (FLS)-released chemoattractants. AZ inhibited HUVEC tube formation in response to the conditioned medium of RA FLS and reduced subintestinal vessel growth in zebrafish, highlighting its anti-angiogenic effects in vitro and in vivo in zebrafish. In AIA rats, AZ mitigated the disease severity and attenuated synovial angiogenesis, as evidenced by decreased numbers of blood vessels and CD31-positive cells. Mechanistically, AZ disrupted the AQP1-FAK interaction, resulting in FAK destabilization and degradation, which was supported by the abrogation of AZ's effects on HUVECs after FAK overexpression. Expectedly, AZ inactivated the FAK-PI3K/Akt pathway in VEGF-induced HUVECs and AIA rat synovium. Collectively, AZ as an AQP1 inhibitor showed anti-rheumatoid effects by reducing angiogenesis through modulation of the FAK-PI3K/AKT pathway.
Collapse
Affiliation(s)
- Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xue-Na Gong
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Yan-Xin Zhou
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Hong-Yan Feng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Min Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei 230032, Anhui Province, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230026, Anhui Province, China.
| | - Li Cai
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China; Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China.
| |
Collapse
|
2
|
Modak D, Ghosh S, Sarkar S, Roy SK, Chakraborty A, Ray A, Patel CN, Georrge JJ, Thakur S, Bhattacharjee S. Unveiling the mechanism of amelioration of adjuvant-induced rheumatoid arthritis by Drynaria quercifolia rhizome extract using network pharmacology and gene expression-based studies. Sci Rep 2025; 15:11981. [PMID: 40199969 PMCID: PMC11978801 DOI: 10.1038/s41598-025-87461-2] [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: 08/08/2024] [Accepted: 01/20/2025] [Indexed: 04/10/2025] Open
Abstract
Rhizomes of Drynaria quercifolia have long been traditionally used to manage rheumatic pain. However, there is limited research supporting this traditional practice and insufficient evidence demonstrating the molecular mechanisms of action of plant-derived bioactives in rheumatoid arthritis (RA). The current study aims to identify the effective components in Drynaria quercifolia methanol rhizome extract (DME) and their probable pharmacological mechanisms in alleviating Rheumatoid Arthritis (RA) using network-pharmacology, molecular docking, molecular-dynamics simulations, and gene expression-based validation. Gas chromatography-mass spectrometry (GC-MS) based screening identified 41 volatile phytocomponents from DME having drug-like potentiality. Network pharmacology-based screening revealed 117 therapeutic targets for RA of which 11 have been identified as core targets. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated that key target genes were mostly enriched in the inflammatory response associated with multiple signalling pathways. Molecular docking and molecular dynamics studies revealed that key target proteins like serine/threonine-protein kinase (AKT1), peroxisome proliferator-activated receptor alpha (PPARA), and peroxisome proliferator-activated receptor gamma (PPARG), exhibited strong binding affinity and stable interactions with multiple phytocomponents present in DME. For experimental verification FCA (Freund's complete adjuvant)-induced chronic arthritis model employed for further molecular investigation. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) results validated that DME significantly (p ≤ 0.05) regulate the expression of key identified target genes AKT and PPARG in experimental RA model. Moreover, this study further confirmed that DME significantly (p ≤ 0.05) downregulated pro-inflammatory mediators like COX-2, IL-6 and TNF-α at gene and protein levels and also normalized (p ≤ 0.05) different oxidative stress parameters in both the low and high dose groups of DME-treated arthritic animals. In conclusion, the network-based in silico approach indicated that the phytocomponents present in DME probably act in a synergistic way to modulate key identified targets associated with RA, which was further validated by experimental studies. Therefore, DME could be a potential alternative in immunomodulatory therapies to combat RA and related chronic inflammatory conditions.
Collapse
Affiliation(s)
- Debabrata Modak
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Subhajit Ghosh
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Sourav Sarkar
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Sudipta K Roy
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Ayan Chakraborty
- Virus Research and Diagnostic Laboratory, North Bengal Medical College and Hospital, Darjeeling, 734012, West Bengal, India
| | - Arpita Ray
- Genetics and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Darjeeling, 734013, West Bengal, India
| | - Chirag N Patel
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Biotechnology Research Center, Technology Innovation Institute, Abu Dhabi, United Arab Emirates
| | - John J Georrge
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India
| | - Subarna Thakur
- Department of Bioinformatics, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India.
| | - Soumen Bhattacharjee
- Cell and Molecular Biology Laboratory, Department of Zoology, University of North Bengal, Raja Rammohunpur, Darjeeling, 734013, West Bengal, India.
| |
Collapse
|
3
|
Huang Y, Zhou MY, Li LL, Lv M, Xu ZS, Wu XJ, Gu SL, Zhang MY, Cai L, Li R. Bacopaside I, acting as an aquaporin 1 inhibitor, ameliorates rheumatoid arthritis via suppressing aquaporin 1-mediated autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156444. [PMID: 39892307 DOI: 10.1016/j.phymed.2025.156444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/16/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Aquaporin 1 (AQP1) is a promising target for regulating fibroblast-like synoviocyte (FLS) behaviors in rheumatoid arthritis (RA). Bacopaside I (BSI), the main active compound of the herbal medicine Bacopa monnieri with anti-RA effects, inhibits tumor cell growth by blocking AQP1, but its potential use in RA is unclear. PURPOSE To address BSI's anti-RA effects and elucidate its underlying mechanisms. METHODS We investigated BSI's therapeutic effects on TNF-α-induced RA FLS and identified AQP1 as its direct target through molecular docking, cellular thermal shift assay (CETSA), and AQP1 knockdown experiments. We studied BSI's impacts on rat adjuvant-induced arthritis (AIA) and synovial proliferation, apoptosis, and autophagy in AIA rat synovium. We explored the role of autophagy inhibition in BSI's effects in vitro and in vivo by co-treating with the autophagy activator rapamycin (Rapa) and/or the inhibitor 3-methyladenine (3-MA). RESULTS BSI suppressed proliferation, promoted apoptosis, and reduced autophagy in TNF-α-stimulated RA FLS. Notably, BSI's in vitro effects were reduced by Rapa and enhanced by 3-MA. The molecular docking and CETSA confirmed BSI's binding to AQP1, while AQP1 knockdown invalidated BSI's in vitro effects, further indicating AQP1 as the target of BSI. In vivo, BSI attenuated the severity of rat AIA, alongside reduced synovial proliferation, increased apoptosis, and decreased autophagy within AIA rat synovium. Moreover, Rapa co-treatment negated BSI's effects on synovial proliferation and apoptosis and abolished its anti-AIA activity. CONCLUSIONS BSI, as an AQP1 inhibitor, hindered AQP1-mediated autophagy, causing increased apoptosis, reduced proliferation in RA FLS, and relieved rat AIA symptoms.
Collapse
Affiliation(s)
- Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Min Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Xin-Jie Wu
- The First Clinical Medical College, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China
| | - Li Cai
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, PR China; Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, PR China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, PR China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230026, Anhui Province, PR China.
| |
Collapse
|
4
|
Chen Q, Chen D, Wang S, Huang X, Liang L, Xie T, Lu J. RND1 Induces Ferroptosis to Alleviate Inflammatory Response, Proliferation, Invasion, and Migration of Rheumatoid Synoviocytes. J Inflamm Res 2025; 18:2647-2659. [PMID: 40008082 PMCID: PMC11853921 DOI: 10.2147/jir.s500630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background Ferroptosis is involved in the occurrence and development of inflammatory arthritis. RND1 has been reported to possess pro-ferroptosis activity. Objective This study was designed to explore the role and the molecular mechanism of RND1 in rheumatoid arthritis (RA). Methods DBA/1 mice were exposed to type II collagen immunization. The pathological damage of the knee joints of mice was observed with H&E staining and RND1 expression in synovial tissues was detected using Western blot. In vitro, Western blot was used to measure RND1, ferroptosis-, migration- and inflammation-related proteins. The cell proliferation, migration and invasion were detected using CCK-8 method, EdU staining, wound healing and transwell assays. The levels of inflammatory factors were detected with ELISA and RT-qPCR. Relative iron level, GSH and MDA concentrations were detected with corresponding assay kits. BODIPY 581/591 C11 kit measured lipid ROS. 4-HNE and GPX4 expression were detected using immunofluorescence assay. Results This study found that RND1 expression was reduced in the synovial tissues of RA mice and human fibroblast-like MH7A synoviocytes. It was also found that the upregulation of RND1 inhibited the proliferation, migration, invasion and inflammatory response in rheumatoid synovial cells via ferroptosis. Conclusion Collectively, RND1 exerted protective impacts on RA, which might be mediated by ferroptosis.
Collapse
Affiliation(s)
- Qiuhua Chen
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Donglan Chen
- Department of Infectious Diseases and Tropical Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Sijie Wang
- Biomedical Diagnostic Center of Ultrastructure, Clinical Research and Experimental Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Xiaomei Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Liang Liang
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Tong Xie
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| | - Jie Lu
- Department of Rheumatology and Immunology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People’s Republic of China
| |
Collapse
|
5
|
Zhang MY, Wang MQ, Huang Y, Gu SL, Zhou MY, Xu ZS, Li LL, Lv M, Cai L, Li R. Silencing aquaporin 1 inhibits autophagy to exert anti-rheumatoid arthritis effects in TNF-α-induced fibroblast-like synoviocytes and adjuvant-induced arthritis rats. Inflamm Res 2025; 74:12. [PMID: 39774992 DOI: 10.1007/s00011-024-01966-6] [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: 10/12/2024] [Revised: 10/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE Fibroblast-like synoviocytes (FLS) are key players in rheumatoid arthritis (RA) by resisting apoptosis via increased autophagy. Elevated synovial aquaporin 1 (AQP1) affects RA FLS behaviors, but its relationship with FLS autophagy is unclear. We aim to clarify that silencing AQP1 inhibits autophagy to exert its anti-RA effects. METHODS We studied the effects and mechanisms of AQP1 silencing on autophagy in TNF-α-induced RA FLS and examined the crucial role of autophagy inhibition in its impacts on RA FLS pathogenic behaviors. We explored whether silencing synovial AQP1 relieved rat adjuvant-induced arthritis (AIA) by reducing synovial autophagy. RESULTS TNF-α stimulation increased AQP1 expression and autophagy levels in RA FLS, with a positive correlation between them. AQP1 silencing inhibited autophagy in TNF-α-stimulated RA FLS, along with suppressing proliferation, promoting apoptosis, and mitigating inflammation. Notably, the inhibitory effects of AQP1 silencing on RA FLS pathogenic behaviors were cancelled by autophagy activation with rapamycin (Rapa) but enhanced by autophagy inhibition using 3-Methyladenine. Mechanistically, silencing AQP1 enhanced the binding of Bcl-2 to Beclin1 by decreasing Beclin1-K63 ubiquitination, thus inhibiting RA FLS autophagy. In vivo, silencing synovial AQP1 relieved the severity and development of rat AIA, alongside reducing Ki67 expression, promoting apoptosis, and decreasing autophagy within AIA rat synovium. Expectedly, the Rapa co-administration nullified the anti-AIA effects of silencing synovial AQP1. CONCLUSION These findings reveal that silencing AQP1 inhibits RA FLS pathogenic behaviors and attenuates rat AIA through autophagy inhibition. This study may help clarify the pathogenic role of AQP1 in enhancing autophagy during RA development.
Collapse
Affiliation(s)
- Man-Yu Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Meng-Qing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Yan Huang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Sheng-Long Gu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Meng-Yuan Zhou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ze-Shan Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ling-Ling Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Min Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Li Cai
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, Anhui Province, China.
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui Province, China.
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230026, Anhui Province, China.
- School of Pharmacy, Anhui Medical University, Hefei, 230032, Anhui Province, China.
| |
Collapse
|
6
|
Zhao Y, Wu Z, Zhou Y, Chen C, Lu Y, Wang H, Xu T, Yang C, Chen X. Cell Sheets Formation Enhances Therapeutic Effects of Human Umbilical Cord Mesenchymal Stem Cells on Spinal Cord Injury. CNS Neurosci Ther 2024; 30:e70163. [PMID: 39670537 PMCID: PMC11638885 DOI: 10.1111/cns.70163] [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: 07/20/2024] [Revised: 10/04/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND In recent years, the utilization of stem cell therapy and cell sheet technology has emerged as a promising approach for addressing spinal cord injury (SCI). However, the most appropriate cell type and mechanism of action remain unclear at this time. This study sought to develop an SCI rat model and evaluate the therapeutic effects of human umbilical cord mesenchymal stem cell (hUC-MSC) sheets in this model. Furthermore, the mechanisms underlying the vascular repair effect of hUC-MSC sheets following SCI were investigated. METHODS A temperature-responsive cell culture method was employed for the preparation of hUC-MSC sheets. The extracellular matrix (ECM) produced by hUC-MSCs serves two distinct yet interrelated purposes. Firstly, it acts as a biologically active scaffold for transplanted cells, facilitating their attachment and proliferation. Secondly, it provides mechanical support and bridges spinal cord stumps, thereby facilitating the restoration of spinal cord function. The formation of the cavity within the spinal cord was evaluated using the Hematoxylin and Eosin (H&E) staining method. Subsequently, endothelial cells were cultivated with the conditioned medium (CM) obtained from hUC-MSCs or hUC-MSC sheets. The pro-angiogenic impact of the conditioned medium of hUC-MSCs (MSC-CM) and the conditioned medium of hUC-MSC sheets (CS-CM) was evaluated through the utilization of the CCK-8 assay, endothelial wound healing assay, and tube formation assay in an in vitro context. The development of glial scars, blood vessels, neurons, and axons in hUC-MSCs and hUC-MSC sheets was assessed through immunofluorescence staining. RESULTS In comparison to hUC-MSCs, hUC-MSC sheets demonstrated a more pronounced capacity to facilitate vascular formation and induce the regeneration of newborn neurons at the SCI site, while also reducing glial scar formation and significantly enhancing motor function in SCI rats. Notably, under identical conditions, the formation of cell sheets has been associated with a paracrine increase in the ability of the cells themselves to secrete pro-angiogenic growth factors. During the course of the experiment, it was observed that the secretion of uPAR was the most pronounced among the pro-angiogenic factors present in MSC-CM and CS-CM. This finding was subsequently corroborated in subsequent experiments, wherein uPAR was demonstrated to promote angiogenesis via the PI3K/Akt signaling pathway. CONCLUSION The creation of cell sheets not only significantly enhances the biological function of hUC-MSCs but also effectively retains the cells locally in spinal cord injury. Therefore, the transplantation of hUC-MSC sheets can maximize the function of hUC-MSCs, greatly reducing glial scar formation, enhancing vascular formation, and promoting the regeneration of neurons and axons. Additionally, the research findings prove that hUC-MSC sheets activate the PI3K/Akt signaling pathway through uPAR secretion to enhance angiogenesis. The transfer of the entire extracellular matrix by hUC-MSC sheets, in the absence of the introduction of additional exogenous or synthetic biomaterials, serves to further augment their potential for clinical application.
Collapse
Affiliation(s)
- Yulin Zhao
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
- Medical School of Nantong UniversityNantongChina
| | - Zhengchao Wu
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
- Medical School of Nantong UniversityNantongChina
| | - Yuchen Zhou
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
- Medical School of Nantong UniversityNantongChina
| | - Cheng Chen
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Yang Lu
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Heng Wang
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Tao Xu
- Medical School of Nantong UniversityNantongChina
| | - Changwei Yang
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| | - Xiaoqing Chen
- Department of Spine SurgeryAffiliated Hospital of Nantong UniversityNantongChina
| |
Collapse
|
7
|
Shi Y, Lei Y, Guo S, Li L, Li X, Liu X, Ding S. Screening Anti-Rheumatoid Arthritis Synovitis Effective Ingredients of Total Flavonoid From Artemisia argyi Folium Based on Spectrum-Effect Relationship. PHYTOCHEMICAL ANALYSIS : PCA 2024. [PMID: 39532485 DOI: 10.1002/pca.3479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Flavonoids are the main nonvolatile component responsible for the anti-rheumatoid arthritis (RA) synovitis activities of Artemisia argyi Folium. However, the effective ingredient remains unidentified. Spectrum-effect relationships analysis was a reliable and efficient strategy for herbal effective ingredient discovery. OBJECTIVES This study aimed to screen the effective ingredients within the total flavonoid from Artemisia argyi Folium (TFAA) that exhibit anti-RA synovitis activities based on spectrum-effect relationship. METHODS TFAA was obtained through ethanol extraction and subsequent purification with D101 resin from 15 distinct batches of Artemisia argyi Folium. The fingerprint of TFAA was established using HPLC, and its efficacy against RA synovitis was evaluated by determining the inhibition rate of nitric oxide (NO) on MH7A synovioblast induced by TNF-α. Common peaks were identified using HPLC-MS/MS and authentic standards. The spectrum-effect relationships between the fingerprints and efficacy were analyzed by gray relational analysis (GRA), canonical correlation analysis (CCA), and partial least squares regression analysis (PLSR) to pinpoint the peaks responsible for the anti-RA synovitis activity, and the results were further verified by in vitro anti-RA synovitis experiments and molecular docking studies. RESULTS The fingerprint revealed 14 common peaks, and 12 compounds were identified, including four caffeoylquinic acids and eight flavonoids. Among them, five flavonoids-X10 (hispidulin), X11 (jaceosidin), X12 (centaureidin), X13 (eupatilin), and X14 (casticin)-were highly relevant to anti-RA synovitis activity. Verification experiments confirmed their inhibitory effect on NO production and cytokine secretion in MH7A cells, showing anti-RA synovitis potential, which was consistent with the established spectrum effect relationship. The underlying mechanism might be related to the inhibition of iNOS. CONCLUSION Hispidulin, jaceosidin, centaureidin, eupatilin, and casticin were the key effective ingredient of TFAA responsible for its anti-RA synovitis effects. These compounds can serve as quality control markers for Artemisia argyi Folium and as lead compounds for anti-RA synovitis treatment.
Collapse
Affiliation(s)
- Yu Shi
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Yueyue Lei
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Shiwen Guo
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Lujun Li
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
- Gansu Herbal Medicine Planting Co., Ltd, Lanzhou, China
| | - Xu Li
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Xinyi Liu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Shuiping Ding
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Qin Z, Tang R, Liang J, Jia X. Berberine, a natural alkaloid: Advances in its pharmacological effects and mechanisms in the treatment of autoimmune diseases. Int Immunopharmacol 2024; 137:112422. [PMID: 38880024 DOI: 10.1016/j.intimp.2024.112422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024]
Abstract
The rising prevalence of autoimmune diseases poses a significant challenge to global public health. Continual exploration of natural compounds for effective treatments for autoimmune diseases is crucial. Berberine, a benzylisoquinoline alkaloid, is a bioactive component found in various medicinal plants, exhibiting diverse pharmacological properties. This review aims to consolidate the current understanding of berberine's pharmacological effects and mechanisms in addressing four autoimmune diseases: rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and psoriasis. Overall, as a traditional Chinese medicinal preparation, berberine shows promise as an effective and safe treatment for autoimmune diseases. However, further comprehensive studies, particularly clinical trials, are essential to elucidate additional mechanisms and molecular targets, as well as to assess the efficacy and safety of berberine in treating these autoimmune diseases.
Collapse
Affiliation(s)
- Zhifang Qin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei Anhui 230012, China
| | - Ran Tang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei Anhui 230012, China
| | - Juan Liang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei Anhui 230012, China
| | - Xiaoyi Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei Anhui 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei Anhui 230012, China.
| |
Collapse
|
9
|
Zou YX, Xiang TN, Xu LR, Zhang H, Ma YH, Zhang L, Zhou CX, Wu X, Huang QL, Lei B, Mu JW, Qin XY, Jiang X, Zheng YJ. Dehydrozaluzanin C- derivative protects septic mice by alleviating over-activated inflammatory response and promoting the phagocytosis of macrophages. Int Immunopharmacol 2024; 132:111889. [PMID: 38531202 DOI: 10.1016/j.intimp.2024.111889] [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: 02/01/2024] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024]
Abstract
Host-directed therapy (HDT) is a new adjuvant strategy that interfere with host cell factors that are required by a pathogen for replication or persistence. In this study, we assessed the effect of dehydrozaluzanin C-derivative (DHZD), a modified compound from dehydrozaluzanin C (DHZC), as a potential HDT agent for severe infection. LPS-induced septic mouse model and Carbapenem resistant Klebsiella pneumoniae (CRKP) infection mouse model was used for testing in vivo. RAW264.7 cells, mouse primary macrophages, and DCs were used for in vitro experiments. Dexamethasone (DXM) was used as a positive control agent. DHZD ameliorated tissue damage (lung, kidney, and liver) and excessive inflammatory response induced by LPS or CRKP infection in mice. Also, DHZD improved the hypothermic symptoms of acute peritonitis induced by CRKP, inhibited heat-killed CRKP (HK-CRKP)-induced inflammatory response in macrophages, and upregulated the proportions of phagocytic cell types in lungs. In vitro data suggested that DHZD decreases LPS-stimulated expression of IL-6, TNF-α and MCP-1 via PI3K/Akt/p70S6K signaling pathway in macrophages. Interestingly, the combined treatment group of DXM and DHZD had a higher survival rate and lower level of IL-6 than those of the DXM-treated group; the combination of DHZD and DXM played a synergistic role in decreasing IL-6 secretion in sera. Moreover, the phagocytic receptor CD36 was increased by DHZD in macrophages, which was accompanied by increased bacterial phagocytosis in a clathrin- and actin-dependent manner. This data suggests that DHZD may be a potential drug candidate for treating bacterial infections.
Collapse
Affiliation(s)
- Ying-Xiang Zou
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tian-Nan Xiang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Chinese Medicine, Hubei College of Chinese Medicine, Jingzhou, Hubei, 434020, China
| | - Li-Rong Xu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huan Zhang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu-He Ma
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lu Zhang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chun-Xian Zhou
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Wu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qi-Lin Huang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Biao Lei
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing-Wen Mu
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiang-Yang Qin
- Department of Chemistry, school of pharmacy, Fourth Military University, Xi'an, Shaanxi 710032, China.
| | - Xin Jiang
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yue-Juan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Health Identification and Assessment, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
10
|
Chen J, Shi X, Deng Y, Dang J, Liu Y, Zhao J, Liang R, Zeng D, Wu W, Xiong Y, Yuan J, Chen Y, Wang J, Lin W, Chen X, Huang W, Olsen N, Pan Y, Fu Q, Zheng SG. miRNA-148a-containing GMSC-derived EVs modulate Treg/Th17 balance via IKKB/NF-κB pathway and treat a rheumatoid arthritis model. JCI Insight 2024; 9:e177841. [PMID: 38652539 PMCID: PMC11141912 DOI: 10.1172/jci.insight.177841] [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: 11/22/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have demonstrated potent immunomodulatory properties that have shown promise in the treatment of autoimmune diseases, including rheumatoid arthritis (RA). However, the inherent heterogeneity of MSCs triggered conflicting therapeutic outcomes, raising safety concerns and limiting their clinical application. This study aimed to investigate the potential of extracellular vesicles derived from human gingival mesenchymal stem cells (GMSC-EVs) as a therapeutic strategy for RA. Through in vivo experiments using an experimental RA model, our results demonstrate that GMSC-EVs selectively homed to inflamed joints and recovered Treg and Th17 cell balance, resulting in the reduction of arthritis progression. Our investigations also uncovered miR-148a-3p as a critical contributor to the Treg/Th17 balance modulation via IKKB/NF-κB signaling orchestrated by GMSC-EVs, which was subsequently validated in a model of human xenograft versus host disease (xGvHD). Furthermore, we successfully developed a humanized animal model by utilizing synovial fibroblasts obtained from patients with RA (RASFs). We found that GMSC-EVs impeded the invasiveness of RASFs and minimized cartilage destruction, indicating their potential therapeutic efficacy in the context of patients with RA. Overall, the unique characteristics - including reduced immunogenicity, simplified administration, and inherent ability to target inflamed tissues - position GMSC-EVs as a viable alternative for RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Jingrong Chen
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Shi
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanan Deng
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junlong Dang
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Liu
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jun Zhao
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongzhen Liang
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | - Yiding Xiong
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Yuan
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ye Chen
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Julie Wang
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weidong Lin
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangfang Chen
- Department of Endocrinology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Nancy Olsen
- Division of Rheumatology, Department of Medicine, The Penn State University Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Yunfeng Pan
- Department of Internal Medicine, Division of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qingling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Song Guo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Zhao J, Liu Y, Shi X, Dang J, Liu Y, Li S, Cai W, Hou Y, Zeng D, Chen Y, Yuan J, Xiong Y, Wu W, Cai P, Chen J, Sun J, Shao Y, Brand DD, Zheng SG. Infusion of GMSCs relieves autoimmune arthritis by suppressing the externalization of neutrophil extracellular traps via PGE2-PKA-ERK axis. J Adv Res 2024; 58:79-91. [PMID: 37169220 PMCID: PMC10982864 DOI: 10.1016/j.jare.2023.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/21/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a systemic autoimmune disease with limited treatment success, characterized by chronic inflammation and progressive cartilage and bone destruction. Accumulating evidence has shown that neutrophil extracellular traps (NETs) released by activated neutrophils are important for initiating and perpetuating synovial inflammation and thereby could be a promising therapeutic target for RA. K/B × N serum transfer-induced arthritis (STIA) is a rapidly developed joint inflammatory model that somehow mimics the inflammatory response in patients with RA. Human gingival-derived mesenchymal stem cells (GMSCs) have been previously shown to possess immunosuppressive effects in arthritis and humanized animal models. However, it is unknown whether GMSCs can manage neutrophils in autoimmune arthritis. OBJECTIVES To evaluate whether infusion of GMSCs can alleviate RA by regulating neutrophils and NETs formation. If this is so, we will explore the underlying mechanism(s) in an animal model of inflammatory arthritis. METHODS The effects of GMSCs on RA were assessed by comparing the symptoms of the K/B × N serum transfer-induced arthritis (STIA) model administered either with GMSCs or with control cells. Phenotypes examined included clinical scores, rear ankle thickness, paw swelling, inflammation, synovial cell proliferation, and immune cell frequency. The regulation of GMSCs on NETs was examined through immunofluorescence and immunoblotting in GMSCs-infused STIA mice and in an in vitro co-culture system of neutrophils with GMSCs. The molecular mechanism(s) by which GMSCs regulate NETs was explored both in vitro and in vivo by silencing experiments. RESULTS We found in this study that adoptive transfer of GMSCs into STIA mice significantly ameliorated experimental arthritis and reduced neutrophil infiltration and NET formation. In vitro studies also showed that GMSCs inhibited the generation of NETs in neutrophils. Subsequent investigations revealed that GMSCs secreted prostaglandin E2 (PGE2) to activate protein kinase A (PKA), which ultimately inhibited the downstream extracellular signal-regulated kinase (ERK) pathway that is essential for NET formation. CONCLUSION Our results demonstrate that infusion of GMSCs can ameliorate inflammatory arthritis mainly by suppressing NET formation via the PGE2-PKA-ERK signaling pathway. These findings further support the notion that the manipulation of GMSCs is a promising stem cell-based therapy for patients with RA and other autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yan Liu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoyi Shi
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Junlong Dang
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yu Liu
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin 541000, China
| | - Siwen Li
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Wei Cai
- Department of Neurology, Mental and Neurological Disease Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yuluan Hou
- Division of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Donglan Zeng
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Ye Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jia Yuan
- Division of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yiding Xiong
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Wenbin Wu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Peihong Cai
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jingrong Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Jianbo Sun
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China
| | - Yiming Shao
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China
| | - David D Brand
- The Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38163, United States
| | - Song Guo Zheng
- Department of Clinical Immunology Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China.
| |
Collapse
|
12
|
Jiang Y, Zhong S, Tan H, Fu Y, Lai J, Liu L, Weng J, Chen H, He S. Study on the mechanism of action of Saposhnikovia divaricata and its key phytochemical on rheumatoid arthritis based on network pharmacology and bioinformatics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117586. [PMID: 38104871 DOI: 10.1016/j.jep.2023.117586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saposhnikovia divaricata (Turcz.) Schischk (SD; called "fangfeng" in China) has been widely used in the clinical treatment of rheumatoid arthritis (RA) and has shown well therapeutic effects, but the specific mechanisms of action of its bioactive phytochemicals remain unclear. AIM OF THE STUDY This study aimed to investigate the molecular biological mechanism of SD in treating RA through a pharmacology-based strategy. The SD-specific core ingredient Prangenidin was screened for further in-depth study. MATERIALS AND METHODS The bioactive phytochemicals of SD and potential targets for the treatment of RA were screened by network pharmacology, and phytochemicals-related parameters such as pharmacology, and toxicology were evaluated. The protein interaction network was established to screen the core targets, and the correlation between the core targets and RA was further validated by bioinformatics strategy. Finally, molecular docking of core components and corresponding targets was performed. The in vitro experiments were performed to elucidate the regulation of Prangenidin on MH7A cells and on the PI3K/AKT pathway, and the in vivo therapeutic effect of Prangenidin was validated in collagen-induced arthritis (CIA) mice. RESULTS A total of 18 bioactive phytochemicals and 66 potential target genes intersecting with the screened RA disease target genes were identified from SD. Finally, core ingredients such as wogonin, beta-sitosterol, 5-O-Methylvisamminol, and prangenidin and core targets such as PTGS2, RELA, and AKT1 were obtained. The underlying mechanism of SD in treating RA might be achieved by regulating pathways such as PI3K/AKT, IL-17 pathway, apoptosis, and multiple biological processes to exert anti-inflammatory and immunomodulatory effects. Molecular docking confirmed that all core ingredients and key targets had great docking activity. Prangenidin inhibited viability, migration, and invasion, and induced apoptosis in MH7A cells. Prangenidin also reduced the production of IL-1β, IL-6, IL-8, MMP-1, and MMP-3. Molecular analysis showed that Prangenidin exerts its regulatory effect on MH7A cells by inhibiting PI3K/AKT pathway. Treatment with Prangenidin ameliorated synovial inflammation in the joints of mice with CIA. CONCLUSION Our findings provide insights into the therapeutic effects of SD on RA, successfully predicting the effective ingredients and potential targets, which could suggest a novel theoretical basis for further exploration of its molecular mechanisms. It also revealed that Prangenidin inhibited viability, migration, invasion, cytokine, and MMPs expression, and induced apoptosis in RA FLSs via the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Yong Jiang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Shuxin Zhong
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Huangsheng Tan
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Yuanfei Fu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Juyi Lai
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Lijin Liu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Juanling Weng
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Hanwei Chen
- Department of Radiology, Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou, 511495, China.
| | - Shenghua He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China; Department of Spine Surgery, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China.
| |
Collapse
|
13
|
Wang Q, Shao G, Zhao X, Wong HH, Chin K, Zhao M, Bai A, Bloom MS, Love ZZ, Chu CR, Cheng Z, Robinson WH. Dysregulated fibrinolysis and plasmin activation promote the pathogenesis of osteoarthritis. JCI Insight 2024; 9:e173603. [PMID: 38502232 PMCID: PMC11141881 DOI: 10.1172/jci.insight.173603] [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: 07/14/2023] [Accepted: 03/05/2024] [Indexed: 03/21/2024] Open
Abstract
Joint injury is associated with risk for development of osteoarthritis (OA). Increasing evidence suggests that activation of fibrinolysis is involved in OA pathogenesis. However, the role of the fibrinolytic pathway is not well understood. Here, we showed that the fibrinolytic pathway, which includes plasminogen/plasmin, tissue plasminogen activator, urokinase plasminogen activator (uPA), and the uPA receptor (uPAR), was dysregulated in human OA joints. Pharmacological inhibition of plasmin attenuated OA progression after a destabilization of the medial meniscus in a mouse model whereas genetic deficiency of plasmin activator inhibitor, or injection of plasmin, exacerbated OA. We detected increased uptake of uPA/uPAR in mouse OA joints by microPET/CT imaging. In vitro studies identified that plasmin promotes OA development through multiple mechanisms, including the degradation of lubricin and cartilage proteoglycans and induction of inflammatory and degradative mediators. We showed that uPA and uPAR produced inflammatory and degradative mediators by activating the PI3K, 3'-phosphoinositide-dependent kinase-1, AKT, and ERK signaling cascades and activated matrix metalloproteinases to degrade proteoglycan. Together, we demonstrated that fibrinolysis contributes to the development of OA through multiple mechanisms and suggested that therapeutic targeting of the fibrinolysis pathway can prevent or slow development of OA.
Collapse
Affiliation(s)
- Qian Wang
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Guoqiang Shao
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection
- Department of Radiology, Stanford Bio-X Program, and
| | - Xiaoyi Zhao
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Heidi H Wong
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Kate Chin
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Mackenzie Zhao
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Audrey Bai
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Michelle S Bloom
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Zelda Z Love
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Constance R Chu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopaedic Surgery, Stanford School of Medicine, Stanford, California, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford, Canary Center at Stanford for Cancer Early Detection
- Department of Radiology, Stanford Bio-X Program, and
| | - William H Robinson
- Division of Immunology & Rheumatology, Stanford School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
14
|
Gong J, Zhang X, Khan A, Liang J, Xiong T, Yang P, Li Z. Identification of serum exosomal miRNA biomarkers for diagnosis of Rheumatoid arthritis. Int Immunopharmacol 2024; 129:111604. [PMID: 38320350 DOI: 10.1016/j.intimp.2024.111604] [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: 11/22/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disorder characterized by inflammation-induced joint damage, which can cause lasting disability. Therefore, early diagnosis and treatment of RA are crucial. Herein, we evaluated whether exosomal microRNAs (miRNAs) could be served as promising biomarkers that can accelerate the diagnosis of RA and development of therapies for RA. METHODS First, we performed small RNA sequencing to determine the miRNA profiles of serum exosomes within a screening cohort comprised of 18 untreated active RA patients, along with 18 age and gender-matched healthy controls (HCs). Subsequently, the miRNA profiles were then validated in a training cohort consisting of 24 RA patients and 24 HCs by RT-qPCR. Finally, the selected exosomal miRNAs were validated in a larger cohort comprising 108 RA patients and 103 HCs. The diagnostic efficacy of the exosomal miRNAs was evaluated by receiver operating characteristic (ROC) curve analysis. Biological functions of the miRNAs were determined by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. RESULTS Our results first demonstrated a noteworthy upregulation of three candidate miRNAs (miR-885-5p, miR-6894-3p, and miR-1268a) in the RA patients' serum exosomes compared to HCs. The combination of three miRNAs along with anti- citrullinated peptide antibodies (ACPA) exhibited excellent diagnostic accuracy, yielding an area under the curve (AUC) of 0.963 (95 % CI : 0.941-0.984), sensitivity of 87.96 %, and specificity of 93.20 %. Notably, miR-885-5p exhibited remarkable discriminatory capacity by itself in indistinguishing ACPA- negative RA patients from HCs, with an AUC of 0.993 (95 % CI : 0.978-1.000), sensitivity of 96.67 %, and specificity of 100 %. Moreover, the expression of miR-1268a in the assessment of therapeutic effectiveness displayed significant reduction on 29th day of Methotrexate (MTX) treatment in RA patients. This decreased expression paralleled with trends observed in tender 28-joint count (TJC28), swollen 28-joint count (SJC28), and disease activity score with 28-joint count using C-reactive protein (DAS28-CRP), all of which are indicative of RA disease activity. Finally, predictive analysis indicated that, these three exosomal miRNAs target pivotal signaling molecules involved in inflammatory pathways, thereby demonstrating effective modulation of the immune system. CONCLUSIONS In this study, we successfully demonstrated the promising potential for serum exosomal miRNAs, particularly miR-885-5p, miR-6894-3p and miR-1268a as biomarkers for early diagnosis and prediction of RA for the first time.
Collapse
Affiliation(s)
- Jianmin Gong
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, China
| | - Xiaoshan Zhang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Adeel Khan
- Department of Biotechnology, University of Science and Technology Bannu, Bannu 28100, Pakistan
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Tao Xiong
- College of Life Science, Yangtze University, Jingzhou, Hubei 434025, China.
| | - Ping Yang
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China.
| | - Zhiyang Li
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
15
|
Liu M, Tang Y, Du Y, Zhang J, Hu F, Zou Y, Li Y, Zhu L, He J, Guo J, Li Z. Leukocyte Ig-like receptor A3 facilitates inflammation, migration and invasion of synovial tissue-derived fibroblasts via ERK/JNK activation. Rheumatology (Oxford) 2024; 63:846-855. [PMID: 37462532 DOI: 10.1093/rheumatology/kead359] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/30/2023] [Indexed: 03/03/2024] Open
Abstract
OBJECTIVE Leukocyte Ig-like receptor A3 (LILRA3) is a soluble receptor belongs to the immunoglobulin superfamily. Our previous studies demonstrated that LILRA3 is a common genetic risk for multiple autoimmune diseases, including RA. Functional LILRA3 conferred increased risk of joint destruction in patients with early RA. We undertook this study to further investigate the pathological role of LILRA3 in joint inflammation of RA. METHODS Soluble LILRA3 was measured by ELISA. LILRA3 plasmids were transfected into human fibroblast-like synoviocytes (FLSs) using electroporation. Activation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was determined by western blots. Cytokine transcripts were quantified by real-time PCR. Migratory and invasive capacities of FLSs were evaluated using transwell migration and Matrigel invasion assays. FLS apoptosis was analysed using flow cytometry. Colocalization of LILRA3, LILRB1 and HLA-G in RA-FLSs was visualized by immunofluorescence staining. RESULTS Soluble LILRA3 was specifically expressed in synovial fluid and serum LILRA3 was significantly increased and positively correlated with disease activity/severity in RA patients. LILRA3 induced an increased expression of IL-6, IL-8 and MMP3 in RA-FLSs. In vitro LILRA3 stimulation or overexpression promoted RA-FLS migration and invasion, and enhanced phosphorylation of ERK/JNK. Inhibition of ERK/JNK resulted in suppression of IL-6/IL-8 expression in LILRA3-stimulated RA-FLSs. LILRA3 was co-localized with its homologue LILRB1 and shared ligand HLA-G in RA-FLSs. CONCLUSION The present study provides the first evidence that soluble LILRA3 is a novel proinflammatory mediator involved in synovial inflammation by promoting RA-FLS activation, migration and invasion, probably through the ERK/JNK signalling pathways.
Collapse
Affiliation(s)
- Mengru Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yundi Tang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yan Du
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jing Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yundong Zou
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Yingni Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Lei Zhu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
16
|
Rufino AT, Freitas M, Proença C, Ferreira de Oliveira JMP, Fernandes E, Ribeiro D. Rheumatoid arthritis molecular targets and their importance to flavonoid-based therapy. Med Res Rev 2024; 44:497-538. [PMID: 37602483 DOI: 10.1002/med.21990] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/18/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
Rheumatoid arthritis (RA) is a progressive, chronic, autoimmune, inflammatory, and systemic condition that primarily affects the synovial joints and adjacent tissues, including bone, muscle, and tendons. The World Health Organization recognizes RA as one of the most prevalent chronic inflammatory diseases. In the last decade, there was an expansion on the available RA therapeutic options which aimed to improve patient's quality of life. Despite the extensive research and the emergence of new therapeutic approaches and drugs, there are still significant unwanted side effects associated to these drugs and still a vast number of patients that do not respond positively to the existing therapeutic strategies. Over the years, several references to the use of flavonoids in the quest for new treatments for RA have emerged. This review aimed to summarize the existing literature about the flavonoids' effects on the major pathogenic/molecular targets of RA and their potential use as lead compounds for the development of new effective molecules for RA treatment. It is demonstrated that flavonoids can modulate various players in synovial inflammation, regulate immune cell function, decrease synoviocytes proliferation and balance the apoptotic process, decrease angiogenesis, and stop/prevent bone and cartilage degradation, which are all dominant features of RA. Although further investigation is necessary to determine the effectiveness of flavonoids in humans, the available data from in vitro and in vivo models suggest their potential as new disease-modifying anti-rheumatic drugs. This review highlights the use of flavonoids as a promising avenue for future research in the treatment of RA.
Collapse
Affiliation(s)
- Ana T Rufino
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Carina Proença
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - José M P Ferreira de Oliveira
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Faculty of Agrarian Sciences and Environment, University of the Azores, Açores, Portugal
| |
Collapse
|
17
|
Liu P, Luo J, Zhao L, Fu Q, Chen Y, Li C, Xu J, Yang X. Causal relationship between several autoimmune diseases and renal malignancies: A two-sample mendelian randomization study. PLoS One 2024; 19:e0297861. [PMID: 38422099 PMCID: PMC10903885 DOI: 10.1371/journal.pone.0297861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/05/2024] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Observational studies have shown an association between systemic autoimmune disease (AD) and multiple malignancies. However, due to the difficulty indetermining the temporal nature of the order, their causal relationship remains elusive. Based on pooled data from a large population-wide genome-wide association study (GWAS), this study explores the genetic causality between systemic autoimmune disease and renal malignancy. METHODS We took a series of quality control steps from a large-scale genome-wide association study to select single nucleotide polymorphisms (SNPs) associated with systemic autoimmune disease as instrumental variables(IVs) to analyze genetic causality with renal malignancies. Inverse variance weighting (IVW), MR- Egger, weighted median, simple model and weighted model were used for analysis. The results were mainly based on IVW (Random Effects), followed by sensitivity analysis. Inverse-Variance Weighted(IVW) and MR-Egger were used to test for heterogeneity. MR- Egger is also used for pleiotropic testing. A single SNP analysis was used to identify single nucleotide polymorphisms (SNPs) with potential impact. Odds ratio (OR) and 95% confidence interval (CI) were used to evaluate causality, and sensitivity analysis was performed to evaluate pleiotropy and instrumental validity. RESULTS Acute and subacute iridocylitis (P = 0.006, OR = 1.077), Ankylosing spondylitis (P = 0.002, OR = 1.051), and spondyloarthritis (P = 0.009, OR = 1.073) were positively associated with an increased risk of renal malignancy. Coxarthrosis (P = 0.008, OR = 0.483), Juvenile rheumatism (P = 0.011, OR = 0.897), and Systemic lupus erythematosus (P = 0.014, OR = 0.869) were negatively associated with an increased risk of renal malignancy. The results of sensitivity analysis were consistent without heterogeneity or pleiotropy. CONCLUSION Our study suggests a causal relationship between different systemic autoimmune diseases and renal malignancies. These findings prompt health care providers to take seriously the potential risk of systemic autoimmune disease and provide new insights into the genetics of kidney malignancies.
Collapse
Affiliation(s)
- Puyu Liu
- Department of Clinical Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jihang Luo
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Lanlan Zhao
- Department of Clinical Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qingqing Fu
- Department of Clinical Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yao Chen
- Department of Clinical Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Chengfang Li
- Department of Clinical Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jieyu Xu
- Department of Pathology, Guiqian International General Hospital, Guiyang, China
| | - Xiaorong Yang
- Department of Clinical Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
18
|
Liu Y, Luo X, Chen Y, Dang J, Zeng D, Guo X, Weng W, Zhao J, Shi X, Chen J, Dong B, Zhong S, Ren J, Li Y, Wang J, Zhang J, Sun J, Xu H, Lu Y, Brand D, Zheng SG, Pan Y. Heterogeneous ferroptosis susceptibility of macrophages caused by focal iron overload exacerbates rheumatoid arthritis. Redox Biol 2024; 69:103008. [PMID: 38142586 PMCID: PMC10788633 DOI: 10.1016/j.redox.2023.103008] [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: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023] Open
Abstract
Focal iron overload is frequently observed in patients with rheumatoid arthritis (RA), yet its functional significance remains elusive. Herein, we report that iron deposition in lesion aggravates arthritis by inducing macrophage ferroptosis. We show that excessive iron in synovial fluid positively correlates with RA disease severity as does lipid hyperoxidation of focal monocyte/macrophages. Further study reveals high susceptibility to iron induced ferroptosis of the anti-inflammatory macrophages M2, while pro-inflammatory M1 are less affected. Distinct glutathione peroxidase 4 (GPX4) degradation depending on p62/SQSTM1 in the two cell types make great contribution mechanically. Of note, ferroptosis inhibitor liproxstatin-1 (LPX-1) can alleviate the progression of K/BxN serum-transfer induced arthritis (STIA) mice accompanied with increasing M2 macrophages proportion. We thus propose that the heterogeneous ferroptosis susceptibility of macrophage subtypes as well as consequent inflammation and immune disorders are potential biomarkers and therapeutic targets in RA.
Collapse
Affiliation(s)
- Yan Liu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiqing Luo
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Ye Chen
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Junlong Dang
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Donglan Zeng
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xinghua Guo
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weizhen Weng
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun Zhao
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiaoyi Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jingrong Chen
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China; Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bo Dong
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shuyuan Zhong
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jianhua Ren
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yuhang Li
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Julie Wang
- Division of Rheumatology and Immunology, Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingwen Zhang
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jianbo Sun
- Department of Clinical Research, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong, China
| | - Hanshi Xu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yan Lu
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - David Brand
- The Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN, 38163, United States
| | - Song Guo Zheng
- Division of Rheumatology and Immunology, Department of Immunology, School of Cell and Gene Therapy, Songjiang Research Institute, Shanghai Songjiang District Central Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yunfeng Pan
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Peng X, Li H, Zhu L, Zhao S, Li Z, Li S, DongtingWu, Chen J, Zheng S, Su W. Single-cell sequencing of the retina shows that LDHA regulates pathogenesis of autoimmune uveitis. J Autoimmun 2024; 143:103160. [PMID: 38160538 DOI: 10.1016/j.jaut.2023.103160] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Autoimmune uveitis (AU) is a severe disorder causing poor vision and blindness. However, the cellular dynamics and pathogenic mechanisms underlying retinal injury in uveitis remain unclear. In this study, single-cell RNA sequencing of the retina and cervical draining lymph nodes in experimental autoimmune uveitis mice was conducted to identify the cellular spatiotemporal dynamics and upregulation of the glycolysis-related gene LDHA. Suppression of LDHA can rescue the imbalance of T effector (Teff) cells/T regulator (Treg) cells under inflammation via downregulation of the glycolysis-PI3K signaling circuit and inhibition of the migration of CXCR4+ Teff cells towards retinal tissue. Furthermore, LDHA and CXCR4 are upregulated in the peripheral blood mononuclear cells of Vogt-Koyanagi-Harada patients. The LDHA inhibitor suppresses CD4+ T cell proliferation in humans. Therefore, our data indicate that the autoimmune environment of uveitis regulates Teff cell accumulation in the retina via glycolysis-associated LDHA. Modulation of this target may provide a novel therapeutic strategy for treating AU.
Collapse
Affiliation(s)
- Xuening Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - He Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Lei Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Sichen Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Si Li
- Sun Yat-sen University, Guangzhou 510060, China
| | - DongtingWu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | | | - Songguo Zheng
- Department of Immunology, School of Cell and Gene Therapy, Shanghai Jiaotong University School of Medicine, 201600, Shanghai, China.
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| |
Collapse
|
20
|
Weng W, Liu Y, Hu Z, Li Z, Peng X, Wang M, Dong B, Zhong S, Jiang Y, Pan Y. Macrophage extracellular traps promote tumor-like biologic behaviors of fibroblast-like synoviocytes through cGAS-mediated PI3K/Akt signaling pathway in patients with rheumatoid arthritis. J Leukoc Biol 2024; 115:116-129. [PMID: 37648663 DOI: 10.1093/jleuko/qiad102] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/01/2023] Open
Abstract
Rheumatoid arthritis is an autoimmune disease characterized by synovium hyperplasia and bone destruction. Macrophage extracellular traps are released from macrophages under various stimuli and may generate stable autoantigen-DNA complexes, as well as aggravate autoantibody generation and autoimmune responses. We aimed to investigate the role of macrophage extracellular traps on the biologic behaviors of rheumatoid arthritis fibroblast-like synoviocytes. Synovial tissues and fibroblast-like synoviocytes were obtained from patients with rheumatoid arthritis. Extracellular traps in synovium and synovial fluids were detected by immunofluorescence, immunohistochemistry, and SYTOX Green staining. Cell viability, migration, invasion, and cytokine expression of rheumatoid arthritis fibroblast-like synoviocytes were assessed by CCK-8, wound-healing assay, Transwell assays, and quantitative real-time polymerase chain reaction, respectively. RNA sequencing analysis was performed to explore the underlying mechanism, and Western blot was used to validate the active signaling pathways. We found that extracellular trap formation was abundant in rheumatoid arthritis and positively correlated to anti-CCP. Rheumatoid arthritis fibroblast-like synoviocytes stimulated with purified macrophage extracellular traps demonstrated the obvious promotion in tumor-like biologic behaviors. The DNA sensor cGAS in rheumatoid arthritis fibroblast-like synoviocytes was activated after macrophage extracellular trap stimuli. RNA sequencing revealed that differential genes were significantly enriched in the PI3K/Akt signaling pathway, and cGAS inhibitor RU.521 effectively reversed the promotion of tumor-like biologic behaviors in macrophage extracellular trap-treated rheumatoid arthritis fibroblast-like synoviocytes and downregulated the PI3K/Akt activation. In summary, our study demonstrates that macrophage extracellular traps promote the pathogenically biological behaviors of rheumatoid arthritis fibroblast-like synoviocytes through cGAS-mediated activation of the PI3K/Akt signaling pathway. These findings provide a novel insight into the pathogenesis of rheumatoid arthritis and the mechanisms of macrophages in modulating rheumatoid arthritis fibroblast-like synoviocyte tumor-like behaviors.
Collapse
Affiliation(s)
- Weizhen Weng
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
- Department of Infectious Disease, The Third People's Hospital of Shenzhen, 29 Bulang Road, Longgang district, Shenzhen, China
| | - Yan Liu
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Zuoyu Hu
- Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Baiyun District, Guangzhou, China
| | - Zhihui Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Xiaohua Peng
- Department of Gastroenterology, The Seventh Affiliated Hospital of Sun Yat-Sen University, 628 Zhenyuan Road, Guangming District, Shenzhen, China
| | - Manli Wang
- Medical Research Center, The Eighth Affiliated Hospital of Sun Yat-sen University, 3025 Shennan Road, Futian District, Shenzhen, China
| | - Bo Dong
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Shuyuan Zhong
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Yutong Jiang
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| | - Yunfeng Pan
- Department of Rheumatology, The Third Affiliated Hospital of Sun Yat-sen University, 600 Tianhe Road, Tianhe District, Guangzhou, China
| |
Collapse
|
21
|
Xue L, Wang B, Zhu J, He Q, Huang F, Wang W, Tao L, Wang Y, Xu N, Yang N, Jin L, Zhang H, Gao N, Lei K, Zhang Y, Xiong C, Lei J, Zhang T, Geng Y, Li M. Profiling of differentially expressed circRNAs and functional prediction in peripheral blood mononuclear cells from patients with rheumatoid arthritis. Ann Med 2023; 55:175-189. [PMID: 36661308 PMCID: PMC9870011 DOI: 10.1080/07853890.2022.2156596] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease associated with an increased risk of death, but its underlying mechanisms are not fully understood. Circular RNAs (circRNAs) have recently been implicated in various biological processes. The aim of this study was to investigate the key circRNAs related to RA. METHODS A microarray assay was used to identify the differentially expressed circRNAs (DEcircRNAs) in peripheral blood mononuclear cells (PBMCs) from patients with RA compared to patients with osteoarthritis (OA) and healthy controls. Then, quantitative real-time PCR was applied to verify the DEcircRNAs, and correlations between the levels of DEcircRNAs and laboratory indices were analysed. We also performed extensive bioinformatic analyses including Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genome (KEGG) pathway and potential circRNA-miRNA-mRNA network analyses to predict the function of these DEcircRNAs. RESULTS A total of 35,342 and 6146 DEcircRNAs were detected in RA patients compared to controls and OA patients, respectively. Nine out of the DEcircRNAs in RA were validated by real-time PCR. There were correlations between the levels of DEcircRNAs and some of the laboratory indices. GO analyses revealed that these DEcircRNAs in RA were closely related to cellular protein metabolic processes, gene expression, the immune system, cell cycle, posttranslational protein modification and collagen formation. Functional annotation of host genes of these DEcircRNAs was implicated in several significantly enriched pathways, including metabolic pathways, ECM-receptor interaction, the PI3K-Akt signalling pathway, the AMPK signalling pathway, leukocyte transendothelial migration, platelet activation and the cAMP signalling pathway, which might be responsible for the pathophysiology of RA. CONCLUSIONS The findings of this study may help to elucidate the role of circRNAs in the specific mechanism underlying RA.Key messagesMicroarray assays showed that a total of 35,342 and 6146 DEcircRNAs were detected in RA patients compared to controls and OA patients, respectively.Nine out of the DEcircRNAs in RA were validated by real-time PCR, and the levels of the DEcircRNAs were related to some of the laboratory indices.GO analyses revealed that the DEcircRNAs in RA were closely related to cellular protein metabolic processes, gene expression, the immune system, etc.Functional annotation of host genes of the DEcircRNAs in RA was implicated in several significantly enriched pathways, including metabolic pathways, ECM-receptor interaction, the PI3K-Akt signalling pathway, etc.
Collapse
Affiliation(s)
- Li Xue
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Clinical Research Center for Endemic Disease of Shaanxi Province, Xi'an, China
| | - Biao Wang
- Department of Immunology and Pathogenic Biology, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Jianhong Zhu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Clinical Research Center for Endemic Disease of Shaanxi Province, Xi'an, China
| | - Qian He
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fang Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wei Wang
- Department of Bone and Joint Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Tao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Nan Xu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ni Yang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hua Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ning Gao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ke Lei
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanping Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chaoliang Xiong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Lei
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ting Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Geng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,National Local Joint Engineering Research Centre of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Qin Y, Jin J, Zhang J, Wang H, Liu L, Zhang Y, Ling S, Hu J, Li N, Wang J, Lv C, Yang X. A fully human monoclonal antibody targeting Semaphorin 5A alleviates the progression of rheumatoid arthritis. Biomed Pharmacother 2023; 168:115666. [PMID: 37832409 DOI: 10.1016/j.biopha.2023.115666] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/03/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is the most common chronic autoimmune disease worldwide. Although progress has been made in RA treatment in recent decades, remission cannot be effectively achieved for a considerable proportion of RA patients. Thus, novel potential targets for therapeutic strategies are needed. Semaphorin 5A (SEMA5A) plays a pivotal role in RA progression by facilitating pannus formation, and it is a promising therapeutic target. In this study, we sought to develop an antibody treatment strategy targeting SEMA5A and evaluate its therapeutic effect using a collagen-induced arthritis (CIA) model. We generated SYD12-12, a fully human SEMA5A blocking antibody, through phage display technology. SYD12-12 intervention effectively inhibited angiogenesis and aggressive phenotypes of RA synoviocytes in vitro and dose-dependently inhibited synovial hyperplasia, pannus formation, bone destruction in CIA mice. Notably, SYD12-12 also improved the Treg/Th17 imbalance in CIA mice. We confirmed through immunofluorescence and molecular docking that SYD12-12 integrated with the unique TSP-1 domain of SEMA5A. In conclusion, we developed and characterized a fully human SEMA5A-blocking antibody for the first time. SYD12-12 effectively alleviated disease progression in CIA mice by inhibiting pannus formation and improving the Treg/Th17 imbalance, demonstrating its potential for the RA treatment.
Collapse
Affiliation(s)
- Yang Qin
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayi Jin
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiani Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hui Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Li Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yanwen Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sunwang Ling
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinzhu Hu
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Nuan Li
- Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianguang Wang
- Institute of Autoimmune Diseases, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Chen Lv
- Department of Orthopedics, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xinyu Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Department of Anesthesia and Critical Care, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
23
|
Wu Y, Wu P, Wu R, Li H, Duan Y, Cai C, Liu Z, She P, Zhang D. Simeprevir restores the anti-Staphylococcus activity of polymyxins. AMB Express 2023; 13:122. [PMID: 37917339 PMCID: PMC10622387 DOI: 10.1186/s13568-023-01634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) infection poses a severe threat to global public health due to its high mortality. Currently, polymyxins are mainly used for the treatment of Gram-negative bacterial-related infection, while exhibiting limited antibacterial activities against Staphylococcus aureus (S. aureus). However, the combination of antibiotics with antibiotic adjuvants is a feasible strategy for the hard-treated infection and toxicity reducing. We will investigate the antibacterial activity of simeprevir (SIM), which treated for genotype 1 and 4 chronic hepatitis C, combined with polymyxins against MRSA through high-throughput screening technology. In our study, the synergistic antibacterial effect of SIM and polymyxins against S. aureus in vitro was found by checkerboard assay and time-growth curve. The cytotoxicity of SIM combined with polymyxin B sulfate [PB(S)] or polymyxin E (PE) in vitro was evaluated using CCK-8, human RBC hemolysis and scratch assays. In addition, we investigated the eradication of biofilm formation of S. aureus by biofilm inhibition assay and the killing of persister cells. Moreover, we evaluated the therapeutic effect and in vivo toxicity of the combination against MRSA in murine subcutaneous abscess model. Furthermore, it was preliminarily found that SIM significantly enhanced the destruction of MRSA membrane by SYTOX Green and DISC3(5) probes. In summary, these results reveal that the therapy of SIM combined with polymyxins (especially PE) is promising for the treatment of MRSA infection.
Collapse
Affiliation(s)
- Yuan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Pingyun Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Ruolan Wu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Huilong Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Yao Duan
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Chaoni Cai
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Zixin Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Pengfei She
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Di Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
24
|
Chang YC, Wu CZ, Cheng CW, Chen JS, Chang LC. Redrawing Urokinase Receptor (uPAR) Signaling with Cancer Driver Genes for Exploring Possible Anti-Cancer Targets and Drugs. Pharmaceuticals (Basel) 2023; 16:1435. [PMID: 37895906 PMCID: PMC10610195 DOI: 10.3390/ph16101435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
During tumorigenesis, urokinase (uPA) and uPA receptor (uPAR) play essential roles in mediating pathological progression in many cancers. To understand the crosstalk between the uPA/uPAR signaling and cancer, as well as to decipher their cellular pathways, we proposed to use cancer driver genes to map out the uPAR signaling. In the study, an integrated pharmaceutical bioinformatics approach that combined modulator identification, driver gene ontology networking, protein targets prediction and networking, pathway analysis and uPAR modulator screening platform construction was employed to uncover druggable targets in uPAR signaling for developing a novel anti-cancer modality. Through these works, we found that uPAR signaling interacted with 10 of 21 KEGG cancer pathways, indicating the important role of uPAR in mediating intracellular cancerous signaling. Furthermore, we verified that receptor tyrosine kinases (RTKs) and ribosomal S6 kinases (RSKs) could serve as signal hubs to relay uPAR-mediated cellular functions on cancer hallmarks such as angiogenesis, proliferation, migration and metastasis. Moreover, we established an in silico virtual screening platform and a uPAR-driver gene pair rule for identifying potential uPAR modulators to combat cancer. Altogether, our results not only elucidated the complex networking between uPAR modulation and cancer but also provided a paved way for developing new chemical entities and/or re-positioning clinically used drugs against cancer.
Collapse
Affiliation(s)
- Yu-Ching Chang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114201, Taiwan;
| | - Chung-Ze Wu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110301, Taiwan;
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan
| | - Chao-Wen Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei City 110301, Taiwan;
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei City 114201, Taiwan
| | - Jin-Shuen Chen
- Department of Education and Research, Kaohsiung Veteran General Hospital, Kaohsiung City 813414, Taiwan
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City 114202, Taiwan
| | - Li-Chien Chang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei City 114201, Taiwan;
- School of Pharmacy, National Defense Medical Center, Taipei City 114201, Taiwan
| |
Collapse
|
25
|
Cai J, Cui J, Wang L. S-palmitoylation regulates innate immune signaling pathways: molecular mechanisms and targeted therapies. Eur J Immunol 2023; 53:e2350476. [PMID: 37369620 DOI: 10.1002/eji.202350476] [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: 04/02/2023] [Revised: 05/10/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
S-palmitoylation is a reversible posttranslational lipid modification that targets cysteine residues of proteins and plays critical roles in regulating the biological processes of substrate proteins. The innate immune system serves as the first line of defense against pathogenic invaders and participates in the maintenance of tissue homeostasis. Emerging studies have uncovered the functions of S-palmitoylation in modulating innate immune responses. In this review, we focus on the reversible palmitoylation of innate immune signaling proteins, with particular emphasis on its roles in the regulation of protein localization, protein stability, and protein-protein interactions. We also highlight the potential and challenge of developing therapies that target S-palmitoylation or de-palmitoylation for various diseases.
Collapse
Affiliation(s)
- Jing Cai
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liqiu Wang
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
26
|
Gao X, Huang X, Wang Y, Sun S, Chen T, Gao Y, Zhang X. Global research hotspots and frontier trends of epigenetic modifications in autoimmune diseases: A bibliometric analysis from 2012 to 2022. Medicine (Baltimore) 2023; 102:e35221. [PMID: 37773838 PMCID: PMC10545364 DOI: 10.1097/md.0000000000035221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/23/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Recent studies have shown substantial progress in understanding the association between epigenetics and autoimmune diseases. However, there is a lack of comprehensive bibliometric analysis in this research area. This article aims to present the current status and hot topics of epigenetic research in autoimmune diseases (ADs) from a bibliometric perspective, as well as explore the frontier hotspots and trends in epigenetic studies related to ADs. METHODS This study collected 1870 epigenetic records related to autoimmune diseases from the web of science core collection database, spanning from 2012 to 2022. Analysis of regions, institutions, journals, authors, and keywords was conducted using CiteSpace, VOSviewer, and the R package "bibliometrix" to predict the latest trends in epigenetic research relevant to autoimmune diseases. RESULTS The number of epigenetic publications related to autoimmune diseases has been increasing annually. The United States has played a major role in this field, contributing over 45.9% of publications and leading in terms of publication volume and citation counts. Central South University emerged as the most active institution, contributing the highest number of publications. Frontiers in Immunology is the most popular journal in this field, publishing the most articles, while the Journal of Autoimmunity is the most co-cited journal. Lu QJ is the most prolific author, and Zhao M is the most frequently co-cited author. "Immunology" serves as a broad representative of epigenetic research in ADs. Hot topics in the field of epigenetic modifications associated with autoimmune diseases include "regulatory T cells (Treg)," "rheumatoid arthritis," "epigenetic regulation," "cAMPresponsive element modulator alpha," "cell-specific enhancer," "genetic susceptibility," and "systemic lupus erythematosus." Furthermore, the study discusses the frontiers and existing issues of epigenetic modifications in the development of autoimmune diseases. CONCLUSIONS This study provides a comprehensive overview of the knowledge structure and developmental trends in epigenetic research related to autoimmune diseases over the past 11 years.
Collapse
Affiliation(s)
- Xiang Gao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Xin Huang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yehui Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Sheng Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Tao Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yongxiang Gao
- International Education College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Xiaodan Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
27
|
Zhang C, Chen S, Wang W, Gao Y. YY1 regulates the proliferation and invasion of triple-negative breast cancer via activating PLAUR. Funct Integr Genomics 2023; 23:269. [PMID: 37552345 DOI: 10.1007/s10142-023-01193-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/09/2023]
Abstract
It is well-established that breast cancer is a highly prevalent malignancy among women, emphasizing the need to investigate mechanisms underlying its pathogenesis and metastasis. In this study, the Gene Expression Omnibus (GEO) database was utilized to conduct differential expression analysis in breast cancer and adjacent tissues. Upregulated genes were selected for prognostic analysis of breast cancer. The expression of urokinase plasminogen activator receptor (uPAR), also known as PLAUR, was assessed using RT-qPCR and western blot. Immunofluorescence staining was employed to determine PLAUR localization. Various cellular processes were analyzed, including proliferation, migration, invasion, apoptosis, and cell cycle. Bioinformatics analysis was used to predict transcription factors of PLAUR, which were subsequently validated in a double luciferase reporter gene experiment. Rescue experiments confirmed the impact of PLAUR on the proliferation, apoptosis, and migration of MDA-MB-231 cells. Furthermore, the effects of PLAUR were evaluated in an orthotopic tumor transplantation and lung metastasis nude mouse model. Our findings substantiated the critical involvement of PLAUR in the progression of triple-negative breast cancer (TNBC) in vitro and among TNBC patients with a poor prognosis. Additionally, we demonstrated Yin Yang-1 (YY1) as a notable transcriptional regulator of PLAUR, whose activation could transcriptionally enhance the proliferation and invasion capabilities of TNBC cells. We also identified the downstream mechanism of PLAUR associated with PLAU, focal adhesion kinase (FAK), and AKT. Overall, these findings offer a novel perspective on PLAUR as a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Chao Zhang
- The First Clinical College, Jinan University, Guangzhou, 510630, China
- Department of Oncology Surgery, The First Affiliated Hospital of Bengbu Medical College, No. 287, Changhuai Road, Longzihu Distract, Bengbu, 233004, Anhui, China
| | - Shiyuan Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Wei Wang
- Department of Oncology Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yong Gao
- The First Clinical College, Jinan University, Guangzhou, 510630, China.
- Department of Vascular Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China.
| |
Collapse
|
28
|
Ballonová L, Kulíšková P, Slanina P, Štíchová J, Vlková M, Hakl R, Litzman J, Souček P, Freiberger T. PLAUR splicing pattern in hereditary angioedema patients' monocytes and macrophages. Mol Biol Rep 2023; 50:4975-4982. [PMID: 37086298 DOI: 10.1007/s11033-023-08391-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/17/2023] [Indexed: 04/23/2023]
Abstract
BACKGROUND The PLAUR gene encodes the urokinase-like plasminogen activator receptor (uPAR) and may undergo alternative splicing. Excluding cassette exons 3, 5 and 6 from the transcript results in truncated protein variants whose precise functions have not been elucidated yet. The PLAUR gene is one of several expressed in myeloid cells, where uPAR participates in different cellular processes, including the contact activation system and kallikrein-kinin system, which play an important role in hereditary angioedema (HAE) pathogenesis. A hypothesis about the PLAUR splicing pattern impact on HAE severity was tested. METHODS AND RESULTS The RT-PCR quantified by capillary electrophoresis was used. Although no significant difference in alternative transcript frequency was observed between healthy volunteers and HAE patients, a significant increase in all cassette exon inclusion variants was revealed during monocyte-to-macrophage differentiation. CONCLUSIONS PLAUR alternative splicing in monocytes and macrophages neither was different between HAE patients and healthy controls, nor reflected disease severity. However, the results showed an PLAUR splicing pattern was changing during monocyte-to-macrophage differentiation, but the significance of these changes is unknown and awaits future clarification.
Collapse
Affiliation(s)
- Lucie Ballonová
- Centre of Cardiovascular Surgery and Transplantation, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petra Kulíšková
- Centre of Cardiovascular Surgery and Transplantation, Brno, Czech Republic
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Peter Slanina
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Julie Štíchová
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marcela Vlková
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Roman Hakl
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiří Litzman
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Přemysl Souček
- Centre of Cardiovascular Surgery and Transplantation, Brno, Czech Republic.
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Tomáš Freiberger
- Centre of Cardiovascular Surgery and Transplantation, Brno, Czech Republic
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| |
Collapse
|
29
|
Sun W, Mao X, Wu W, Nan Y, Xu C, Wang Y, Xu H. Inhibition of Cdc37 Ameliorates Arthritis in Collagen-Induced Arthritis Rats by Inhibiting Synoviocyte Proliferation and Migration Through the ERK Pathway. Inflammation 2023; 46:1022-1035. [PMID: 36920636 DOI: 10.1007/s10753-023-01789-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/18/2022] [Accepted: 02/01/2023] [Indexed: 03/16/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that can lead to synovial inflammation, pannus formation, cartilage damage, bone destruction, and ultimate disability. Fibroblast-like synoviocytes (FLS) are involved in the pathogenetic mechanism of RA. Cdc37 (cell division cycle protein 37) is regarded as a molecular chaperone involved in various physiological processes such as cell cycle progression, cell proliferation, cell signal transduction, tumorigenesis, and progression. However, the precise role of Cdc37 in the pathogenesis of rheumatoid arthritis (RA) remains uncertain. In our study, we found that Cdc37 expression was upregulated in human rheumatoid synovia in contrast with the normal group. Interestingly, Cdc37 activated the ERK pathway to promote RA-FLS proliferation and migration in vitro. Ultimately, in vivo experiments revealed that silencing of Cdc37 alleviated ankle swelling and cartilage destruction and validated the ERK signaling pathways in vitro findings. Collectively, we demonstrate that Cdc37 promotes the proliferation and migration of RA-FLS by activation of ERK signaling pathways and finally aggravates the progression of RA. These data indicated that Cdc37 may be a novel target for the treatment of RA.
Collapse
Affiliation(s)
- Weiwei Sun
- Department of Orthopaedics, Medical School, Affiliated Hospital of Nantong University, Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Xingxing Mao
- Department of Orthopaedics, Affiliated Nantong Hospital of Shanghai University, The Six People's Hospital of Nantong, Jiangsu, 226001, Nantong, China
| | - Weijie Wu
- Department of Orthopaedics, Medical School, Affiliated Hospital of Nantong University, Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu, China.,Department of Orthopaedics, Affiliated Nantong Hospital of Shanghai University, The Six People's Hospital of Nantong, Jiangsu, 226001, Nantong, China
| | - Yunyi Nan
- Department of Orthopaedics, Medical School, Affiliated Hospital of Nantong University, Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu, China
| | - Chunxiang Xu
- Department of Nursing, Medical School, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China.
| | - Youhua Wang
- Department of Orthopaedics, Medical School, Affiliated Hospital of Nantong University, Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| | - Hua Xu
- Department of Orthopaedics, Medical School, Affiliated Hospital of Nantong University, Nantong University, No. 20 Xisi Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
30
|
Benucci M, Damiani A, Russo E, Guiducci S, Li Gobbi F, Fusi P, Grossi V, Amedei A, Manfredi M, Infantino M. The Association of uPA, uPAR, and suPAR System with Inflammation and Joint Damage in Rheumatoid Arthritis: suPAR as a Biomarker in the Light of a Personalized Medicine Perspective. J Pers Med 2022; 12:1984. [PMID: 36556207 PMCID: PMC9788564 DOI: 10.3390/jpm12121984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/10/2022] [Accepted: 11/23/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND In recent years, the involvement of the soluble urokinase Plasminogen Activator Receptor (suPAR) in the pathophysiological modulation of Rheumatoid Arthritis (RA) has been documented, resulting in the activation of several intracellular inflammatory pathways. METHODS We investigated the correlation of urokinase Plasminogen Activator (uPA)/urokinase Plasminogen Activator Receptor (uPAR) expression and suPAR with inflammation and joint damage in RA, evaluating their potential role in a precision medicine context. RESULTS Currently, suPAR has been shown to be a potential biomarker for the monitoring of Systemic Chronic Inflammation (SCI) and COVID-19. However, the effects due to suPAR interaction in immune cells are also involved in both RA onset and progression. To date, the literature data on suPAR in RA endorse its potential application as a biomarker of inflammation and subsequent joint damage. CONCLUSION Available evidence about suPAR utility in the RA field is promising, and future research should further investigate its use in clinical practice, resulting in a big step forward for precision medicine. As it is elevated in different types of inflammation, suPAR could potentially work as an adjunctive tool for the screening of RA patients. In addition, a suPAR system has been shown to be involved in RA pathogenesis, so new data about the therapeutic response to Jak inhibitors can represent a possible way to develop further studies.
Collapse
Affiliation(s)
- Maurizio Benucci
- Rheumatology Unit, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, 50143 Florence, Italy
| | - Arianna Damiani
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Edda Russo
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Serena Guiducci
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Francesca Li Gobbi
- Rheumatology Unit, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, 50143 Florence, Italy
| | - Paola Fusi
- Rheumatology Unit, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, 50143 Florence, Italy
| | - Valentina Grossi
- Immunology and Allergology Laboratory, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, 50143 Florence, Italy
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50134 Florence, Italy
| | - Mariangela Manfredi
- Immunology and Allergology Laboratory, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, 50143 Florence, Italy
| | - Maria Infantino
- Immunology and Allergology Laboratory, Hospital S. Giovanni di Dio, Azienda USL-Toscana Centro, 50143 Florence, Italy
| |
Collapse
|
31
|
Tao SS, Cao F, Sam NB, Li HM, Feng YT, Ni J, Wang P, Li XM, Pan HF. Dickkopf-1 as a promising therapeutic target for autoimmune diseases. Clin Immunol 2022; 245:109156. [DOI: 10.1016/j.clim.2022.109156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/24/2022] [Accepted: 10/06/2022] [Indexed: 11/03/2022]
|
32
|
Yang C, Liu Q, Jiang Z. CircPTTG1IP knockdown suppresses rheumatoid arthritis progression by targeting miR-431-5p/FSTL1 axis. Transpl Immunol 2022; 75:101685. [PMID: 35933079 DOI: 10.1016/j.trim.2022.101685] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 10/16/2022]
Abstract
BACKGROUND It is observed that circular RNA (circRNA) PTTG1 interacting protein (circPTTG1IP) level is notably up-regulated in rheumatoid arthritis (RA) patients by previous study. However, its precise role and working mechanism in RA pathology remain to be clarified. METHODS AND RESULTS Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot assay were carried out to examine RNA and protein expression. Cell proliferation was analyzed by colony formation assay and 5-Ethynyl-2'-deoxyuridine (EdU) assay. Cell motility was assessed by transwell assays and wound healing assay. Flow cytometry (FCM) analysis was performed to assess cell apoptosis rate. Dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA-pull down assays were conducted to confirm the interaction between microRNA-431-5p (miR-431-5p) and circPTTG1IP or follistatin like 1 (FSTL1). CircPTTG1IP expression was up-regulated in the synovial tissues of RA patients and RA patients-derived fibroblast-like synoviocytes (RA-FLS). CircPTTG1IP absence suppressed the proliferation, migration, and invasion and induced the apoptosis of RA-FLS. CircPTTG1IP negatively regulated the expression of miR-431-5p by directly binding to it in RA-FLS. CircPTTG1IP interference-mediated effects in RA-FLS were largely counteracted by the silence of miR-431-5p. miR-431-5p directly interacted with the 3' untranslated region (3'UTR) of FSTL1. FSTL1 overexpression largely overturned miR-431-5p accumulation-mediated effects in RA-FLS. CircPTTG1IP positively regulated FSTL1 expression by sponging miR-431-5p in RA-FLS. CONCLUSION CircPTTG1IP absence suppressed RA progression through mediating miR-431-5p/FSTL1 signaling cascade.
Collapse
Affiliation(s)
- Chenhui Yang
- Department of Orthopedic, Tianshui Hand and Foot of Surgery Hospital, China
| | - Qingling Liu
- Department of Orthopedic, Tianshui Hand and Foot of Surgery Hospital, China.
| | - Zaiming Jiang
- Department of Orthopedic, Tianshui Hand and Foot of Surgery Hospital, China
| |
Collapse
|
33
|
Wang R, Li H, Han Y, Li L. Knockdown of circ_0025908 inhibits proliferation, migration, invasion, and inflammation while stimulates apoptosis in fibroblast-like synoviocytes by regulating miR-650-dependent SCUBE2. Autoimmunity 2022; 55:473-484. [PMID: 35904110 DOI: 10.1080/08916934.2022.2102164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
BACKGROUND Circular RNAs (circRNAs) are demonstrated to play vital roles in human diseases, including rheumatoid arthritis (RA). Therefore, this research aimed to explore the effects of hsa_circRNA_0025908 (circ_0025908) on RA. METHODS RNA expression of circ_0025908, microRNA-650 (miR-650), and Signal peptide-CUBepidermal growth factor-like containing protein 2 (SCUBE2) were assessed by real-time quantitative polymerase chain reaction; protein expression of SCUBE2, apoptosis- and invasion-related proteins was evaluated by western blot assay. Functional assays were performed using 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide, 5-ethynyl-2'-deoxyuridine, transwell, flow cytometry, and enzyme linked immunosorbent assay assays. Dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays confirmed the interaction relationship among circ_0025908, miR-650, and SCUBE2. RESULTS Circ_0025908 was overexpressed in synovial tissues and fibroblast-like synoviocytes (FLS) from RA patients. Inhibition of circ_0025908 repressed proliferation, migration, invasion, inflammation, and cell cycle progression, while induced apoptosis in the FLS isolated from RA patients (FLS-RA), accompanied with increased Bax, cleaved caspase-3 and E-cadherin, but declined Bcl-2, N-cadherin and Vimentin. MiR-650 was a target of circ_0025908, and SCUBE2 was a target for miR-650. Silencing of miR-650 could overturned above effects of circ_0025908 knockdown in FLS-RA, whereas its overexpression could mimic those effects by downregulating SCUBE2. Additionally, SCUBE2 expression could be positively regulated by circ_0025908 and inversely regulated by miR-650. Notably, Pearson's correlation analysis confirmed the linear correlation among circ_0025908, miR-650 and SCUBE2 in these RA tissues. CONCLUSION Circ_0025908 inhibition can suppress FLS-RA dysfunctions through targeting miR-650/SCUBE2 axis, suggesting a new potential therapeutic clue for RA patients.
Collapse
Affiliation(s)
- Ronghua Wang
- Department of Rheumatology and Immunology, Xingtai People's Hospital, Xingtai, Hebei Province, China
| | - Hongbo Li
- Department of Anesthesiology, Weifang People's Hospital, Weifang, China
| | - Yunning Han
- Department of Pain, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lei Li
- Department of Pain, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
34
|
Rheumatoid arthritis fibroblast-like synoviocytes maintain tumor-like biological characteristics through ciRS-7-dependent regulation of miR-7. Mol Biol Rep 2022; 49:8473-8483. [PMID: 35752700 DOI: 10.1007/s11033-022-07666-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/25/2022] [Accepted: 05/31/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Altered phenotype of Fibroblast-like synoviocyte(FLS) is an important cause of the pathogenesis and progression of rheumatoid arthritis(RA), but the specific mechanism causing this change has not yet been fully explained. The exact mechanism by which the biological properties of FLS change in RA is still unclear. microRNAs (miRNAs) have been shown to affect changes in the biological properties of RA-FLS, but the critical miRNAs remain to be discovered. Thus, we first used miRNA microarray and WGCNA to confirm the RA-FLS miRNA landscape and establish their biological functions via network analyses at the system level, as well as to provide a platform for modulating the overall phenotypic effects of RA-FLS. METHODS We enrolled a total of 3 patients with RA and 3 healthy participants, constructed a network analysis of via miRNA microarray and RNA-sequencing. Furthermore, the coexpression analyses of miR-7 and ciRS-7 were verified by siRNA transfection, overexpression and qPCR analyses. Finally, we evaluated the effects of adjusting the expression levels of miR-7 and ciRS-7 on RA-FLS, respectively. RESULTS We identified distinct miRNA features in RA-FLS, including miR-7, which was significantly lower expressed. Furthermore, we discovered the negative regulatory relationship between ciRS-7 and miR-7 in RA-FLS. Finally, we overexpressed miR-7 in RA-FLS and discovered that miR-7 inhibited RA-FLS hyperproliferation, migration, invasion, and apoptosis, whereas ciRS-7 overexpression reversed these effects. CONCLUSIONS The results indicate that the dysregulation of miR-7 in FLS may be involved in the pathological processes of RA and that ciRS-7 induced the suppression of tumor-like biological characters of RA-FLS via modulation of miR-7. These findings help us understand the essential roles of a regulatory interaction between ciRS-7 and miR-7 mediating disease activity of RA, and will facilitate to develop potential intervention target for RA.
Collapse
|
35
|
Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms23137071. [PMID: 35806074 PMCID: PMC9267012 DOI: 10.3390/ijms23137071] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022] Open
Abstract
Extensive angiogenesis is a characteristic feature in the synovial tissue of rheumatoid arthritis (RA) from a very early stage of the disease onward and constitutes a crucial event for the development of the proliferative synovium. This process is markedly intensified in patients with prolonged disease duration, high disease activity, disease severity, and significant inflammatory cell infiltration. Angiogenesis is therefore an interesting target for the development of new therapeutic approaches as well as disease monitoring strategies in RA. To this end, nuclear imaging modalities represent valuable non-invasive tools that can selectively target molecular markers of angiogenesis and accurately and quantitatively track molecular changes in multiple joints simultaneously. This systematic review summarizes the imaging markers used for single photon emission computed tomography (SPECT) and/or positron emission tomography (PET) approaches, targeting pathways and mediators involved in synovial neo-angiogenesis in RA.
Collapse
|
36
|
Sonic Hedgehog Promotes Proliferation and Migration of Fibroblast-Like Synoviocytes in Rheumatoid Arthritis via Rho/ROCK Signaling. J Immunol Res 2022; 2022:3423692. [PMID: 35785032 PMCID: PMC9242744 DOI: 10.1155/2022/3423692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 01/13/2023] Open
Abstract
Objective. To explore the underlying mechanism of the sonic hedgehog (Shh) signaling pathway in promoting cell proliferation and migration in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). Method. FLS were collected from 8 patients with RA and 3 patients with osteoarthritis (OA). The expression of smoothened (Smo, the Shh pathway activator) was quantified by real-time PCR and western blot. FLS were incubated with cyclopamine (a Smo antagonist), purmorphamine (a Smo agonist), Y27632 (a Rho/ROCK signaling inhibitor), or a combination of purmorphamine and Y27632, respectively. Cell proliferation was examined using cell counting kit-8 and cell cycle assays while cell migration was measured with Transwell and wound healing assays. Results. The expression of Smo was higher in FLS from RA patients than from OA patients (
). RA-FLS treated with purmorphamine showed significantly activated proliferation (119.69 vs. 100.0) and migration (252.38 vs. 178.57) compared to untreated cells (both
). RA-FLS incubated with cyclopamine or a combination of purmorphamine and Y27632 exhibited significant suppression of proliferation (81.55 vs. 100.0 and 85.84 vs. 100.0) and migration (100 vs. 178.57 and 109.52 vs. 185) ability (all
). Conclusion. Our results demonstrated that Shh promoted cell growth and migration of FLS in RA patients through the Rho/ROCK signaling pathway.
Collapse
|
37
|
Zhang Y, Yang X, Han C, Wang D, Ma Y, Wei W. Paeoniflorin‑6'O‑benzene sulfonate suppresses fibroblast‑like synoviocytes proliferation and migration in rheumatoid arthritis through regulating GRK2‑Gβγ interaction. Exp Ther Med 2022; 24:523. [DOI: 10.3892/etm.2022.11450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/19/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Yuwen Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti‑inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti‑inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti‑inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti‑inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Chenchen Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti‑inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti‑inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Dandan Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti‑inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti‑inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Yang Ma
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti‑inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti‑inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti‑inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti‑inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
38
|
Xu Z, Shang W, Zhao Z, Zhang B, Liu C, Cai H. Curcumin alleviates rheumatoid arthritis progression through the phosphatidylinositol 3-kinase/protein kinase B pathway: an in vitro and in vivo study. Bioengineered 2022; 13:12899-12911. [PMID: 35609329 PMCID: PMC9276000 DOI: 10.1080/21655979.2022.2078942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease characterized by synovial inflammation and joint bone and cartilage destruction. Curcumin can improve joint inflammation in rats with arthritis and inhibit synovial revascularization and abnormal proliferation of fibroblasts. However, it is unclear whether curcumin affects the RA progression. The TNF-α-stimulated primary RA fibroblast-like synoviocytes (RA-FLS) and SV-40 transformed MH7A cells were used as the in vitro model of RA. A mouse model of collagen-induced arthritis (CIA) was used as the in vivo model. The effects of curcumin on cell proliferation, apoptosis, migration, invasion, and inflammatory response were assessed by colony formation, flow cytometry, wound scratch, Transwell assays, and western blotting analysis. Arthritis index scores and degree of paw swelling in mice were assessed to evaluate RA. Curcumin inhibited the TNF-α-induced proliferation, migration, invasion of MH7A and RA-FLS cells and promoted cell apoptosis. Administration with curcumin reversed the CIA-induced increase in arthritis scores, hind paw edema, and loss of appetite, while these effects were rescued by insulin-like growth factor 1, the upstream cytokine of PI3K/AKT. Moreover, curcumin suppressed the inflammatory response by reducing TNF-α, IL-6, and IL-17 secretion in CIA-stimulated mice. Curcumin has an excellent anti-RA effect in vivo and in vitro, which is exerted by inhibiting the expression of pro-inflammatory factors TNF-a, IL-6 and IL-17 and inhibiting the activation of PI3K/AKT signaling pathway. Thus, curcumin may be a promising candidate for anti-RA treatment.
Collapse
Affiliation(s)
- Zihan Xu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.,Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Wei Shang
- Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Zhiming Zhao
- Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Beibei Zhang
- Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Chunli Liu
- Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Hui Cai
- Department of Integrated Traditional and Western Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
39
|
Ma M, Fan AY, Liu Z, Yang LQ, Huang JM, Pang ZY, Yin F. Baohuoside I Inhibits Osteoclastogenesis and Protects Against Ovariectomy-Induced Bone Loss. Front Pharmacol 2022; 13:874952. [PMID: 35571086 PMCID: PMC9092047 DOI: 10.3389/fphar.2022.874952] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/28/2022] [Indexed: 11/18/2022] Open
Abstract
Bone-resorbing osteoclasts are essential for skeletal remodelling, and the hyperactive formation and function of osteoclasts are common in bone metabolic diseases, especially postmenopausal osteoporosis. Therefore, regulating the osteoclast differentiation is a major therapeutic target in osteoporosis treatment. Icariin has shown potential osteoprotective effects. However, existing studies have reported limited bioavailability of icariin, and the material basis of icariin for anti-osteoporosis is attributed to its metabolites in the body. Here, we compared the effects of icariin and its metabolites (icariside I, baohuoside I, and icaritin) on osteoclastogenesis by high-content screening followed by TRAP staining and identified baohuoside I (BS) with an optimal effect. Then, we evaluated the effects of BS on osteoclast differentiation and bone resorptive activity in both in vivo and in vitro experiments. In an in vitro study, BS inhibited osteoclast formation and bone resorption function in a dose-dependent manner, and the elevated osteoclastic-related genes induced by RANKL, such as NFATc1, cathepsin K, RANK, and TRAP, were also attenuated following BS treatment. In an in vivo study, OVX-induced bone loss could be prevented by BS through interrupting the osteoclast formation and activity in mice. Furthermore, mechanistic investigation demonstrated that BS inhibited osteoclast differentiation by ameliorating the activation of the MAPK and NF-kB pathways and reducing the expression of uPAR. Our study demonstrated that baohuoside I could inhibit osteoclast differentiation and protect bone loss following ovariectomy.
Collapse
Affiliation(s)
- Min Ma
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ao-Yuan Fan
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zheng Liu
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li-Qing Yang
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun-Ming Huang
- Department of Orthopaedic, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhi-Ying Pang
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Feng Yin
- Department of Joint Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China.,Shanghai Clinical Research Centre for Ageing and Medicine, Shanghai, China
| |
Collapse
|
40
|
Chen Y, Dang J, Lin X, Wang M, Liu Y, Chen J, Chen Y, Luo X, Hu Z, Weng W, Shi X, Bi X, Lu Y, Pan Y. RA Fibroblast-Like Synoviocytes Derived Extracellular Vesicles Promote Angiogenesis by miRNA-1972 Targeting p53/mTOR Signaling in Vascular Endotheliocyte. Front Immunol 2022; 13:793855. [PMID: 35350778 PMCID: PMC8957937 DOI: 10.3389/fimmu.2022.793855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/10/2022] [Indexed: 01/20/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammatory in joints. Invasive pannus is a characteristic pathological feature of RA. RA fibroblast-like synoviocytes (FLSs) are showed tumor-like biological characters that facilitate pannus generation. Importantly, it has been documented that extracellular vesicle (EVs) derived microRNAs have a vital role of angiogenesis in various immune inflammatory diseases. However, whether RA FLSs derived EVs can facilitate angiogenesis and the underlying mechanism is undefined. Herein, we aim to investigate the key role of RA FLSs derived EVs on angiogenesis in endothelial cells (ECs). We indicate that RA FLSs derived EVs promote ECs angiogenesis by enhancing migration and tube formation of ECs in vitro. Also, we confirm that RA FLSs derived EVs can significantly facilitate ECs angiogenesis with a matrigel angiogenesis mice model. In terms of the mechanisms, both RNAs and proteins in EVs play roles in promoting ECs angiogenesis, but the RNA parts are more fundamental in this process. By combining microRNA sequencing and qPCR results, miR-1972 is identified to facilitate ECs angiogenesis. The blockage of miR-1972 significantly abrogated the angiogenesis stimulative ability of RA FLSs derived EVs in ECs, while the overexpression of miR-1972 reversed the effect in ECs. Specifically, the p53 level is decreased, and the phosphorylated mTOR is upregulated in miR-1972 overexpressed ECs, indicating that miR-1972 expedites angiogenesis through p53/mTOR pathway. Collectively, RA FLSs derived EVs can promote ECs angiogenesis via miR-1972 targeted p53/mTOR signaling, targeting on RA FLSs derived EVs or miR-1972 provides a promising strategy for the treatment of patients with RA.
Collapse
Affiliation(s)
- Yixiong Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Rheumatology, Affiliated Dongguan People's Hospital, Southern Medical University, Dongguan, China
| | - Junlong Dang
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaorong Lin
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Manli Wang
- Medical Research Center, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yan Liu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingrong Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ye Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiqing Luo
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zuoyu Hu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weizhen Weng
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Shi
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuan Bi
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Lu
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunfeng Pan
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
41
|
In Vitro Characterization of Doxorubicin-Mediated Stress-Induced Premature Senescence in Human Chondrocytes. Cells 2022; 11:cells11071106. [PMID: 35406671 PMCID: PMC8998002 DOI: 10.3390/cells11071106] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023] Open
Abstract
Accumulation of senescent chondrocytes is thought to drive inflammatory processes and subsequent cartilage degeneration in age-related as well as posttraumatic osteoarthritis (OA). However, the underlying mechanisms of senescence and consequences on cartilage homeostasis are not completely understood so far. Therefore, suitable in vitro models are needed to study chondrocyte senescence. In this study, we established and evaluated a doxorubicin (Doxo)-based model of stress-induced premature senescence (SIPS) in human articular chondrocytes (hAC). Cellular senescence was determined by the investigation of various senescence associated (SA) hallmarks including β-galactosidase activity, expression of p16, p21, and SA secretory phenotype (SASP) markers (IL-6, IL-8, MMP-13), the presence of urokinase-type plasminogen activator receptor (uPAR), and cell cycle arrest. After seven days, Doxo-treated hAC displayed a SIPS-like phenotype, characterized by excessive secretion of SASP factors, enhanced uPAR-positivity, decreased proliferation rate, and increased β-galactosidase activity. This phenotype was proven to be stable seven days after the removal of Doxo. Moreover, Doxo-treated hAC exhibited increased granularity and flattened or fibroblast-like morphology. Further analysis implies that Doxo-mediated SIPS was driven by oxidative stress as demonstrated by increased ROS levels and NO release. Overall, we provide novel insights into chondrocyte senescence and present a suitable in vitro model for further studies.
Collapse
|
42
|
Achudhan D, Liu SC, Lin YY, Huang CC, Tsai CH, Ko CY, Chiang IP, Kuo YH, Tang CH. Antcin K Inhibits TNF-α, IL-1β and IL-8 Expression in Synovial Fibroblasts and Ameliorates Cartilage Degradation: Implications for the Treatment of Rheumatoid Arthritis. Front Immunol 2022; 12:790925. [PMID: 34975889 PMCID: PMC8714747 DOI: 10.3389/fimmu.2021.790925] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
Extracts from Taiwan’s traditional medicinal mushroom, Antrodia cinnamomea, exhibit anti-inflammatory activities in cellular and preclinical studies. However, this paper is the first to report that Antcin K, a triterpenoid isolated from A. cinnamomea, inhibits proinflammatory cytokine production in human rheumatoid synovial fibroblasts (RASFs), which are major players in rheumatoid arthritis (RA) disease. In our analysis of the mechanism of action, Antcin K inhibited the expression of three cytokines (tumor necrosis factor alpha [TNF-α], interleukin 1 beta [IL-1β] and IL-8) in human RASFs; cytokines that are crucial to RA synovial inflammation. Notably, incubation of RASFs with Antcin K reduced the phosphorylation of the focal adhesion kinase (FAK), phosphoinositide 3-kinase (PI3K), protein kinase B (AKT) and nuclear factor-κB (NF-κB) signaling cascades, all of which promote cytokine production in RA. Intraperitoneal injections of Antcin K (10 mg/kg or 30 mg/kg) attenuated paw swelling, cartilage degradation and bone erosion, and decreased serum levels of TNF-α, IL-1β, IL-8 in collagen-induced arthritis (CIA) mice; in further experiments, IL-6 levels were similarly reduced. The inhibitory effects of Antcin K upon TNF-α, IL-1β and IL-8 expression in human RASFs was achieved through the downregulation of the FAK, PI3K, AKT and NF-κB signaling cascades. Our data support clinical investigations using Antcin K in RA disease.
Collapse
Affiliation(s)
- David Achudhan
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Yen-You Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Chien-Chung Huang
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung, Taiwan.,Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Yuan Ko
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - I-Ping Chiang
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
43
|
Liu C, Fan F, Zhong L, Su J, Zhang Y, Tu Y. Elucidating the material basis and potential mechanisms of Ershiwuwei Lvxue Pill acting on rheumatoid arthritis by UPLC-Q-TOF/MS and network pharmacology. PLoS One 2022; 17:e0262469. [PMID: 35130279 PMCID: PMC8820630 DOI: 10.1371/journal.pone.0262469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/23/2021] [Indexed: 11/18/2022] Open
Abstract
Ershiwuwei Lvxue Pill (ELP, མགྲིན་མཚལ་ཉེར་ལྔ།), a traditional Tibetan medicine preparation, has been used hundreds of years for the clinical treatment of rheumatoid arthritis (RA) in the highland region of Tibet, China. Nevertheless, its chemical composition and therapeutic mechanism are unclear. This study aimed to uncover the potentially effective components of ELP and the pharmacological mechanisms against RA by combing UPLC-Q-TOF/MS and network pharmacology. In this study, 96 compounds of ELP were identified or tentatively characterized based on UPLC-Q-TOF/MS analysis. Then, a total of 22 potential bioactive compounds were screened by TCMSP with oral bioavailability and drug-likeness. Preliminarily, 10 crucial targets may be associated with RA through protein-protein interaction network analysis. The functional enrichment analysis indicated that ELP exerted anti-RA effects probably by synergistically regulating many biological pathways, such as PI3K-Akt, Cytokine-cytokine receptor interaction, JAK-STAT, MAPK, TNF, and Toll-like receptor signaling pathway. In addition, good molecular docking scores were highlighted between five promising bioactive compounds (ellagic acid, quercetin, kaempferol, galangin, coptisine) and five core targets (PTGS2, STAT3, VEGFA, MAPK3, TNF). Overall, ELP can exert its anti-RA activity via multicomponent, multitarget, and multichannel mechanisms of action. However, further studies are needed to validate the biological processes and effect pathways of ELP.
Collapse
Affiliation(s)
- Chuan Liu
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Fangfang Fan
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Zhong
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinsong Su
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhang
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ya Tu
- Development Research Center of Traditional Chinese Medicine, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Gao Y, Vogus D, Zhao Z, He W, Krishnan V, Kim J, Shi Y, Sarode A, Ukidve A, Mitragotri S. Injectable hyaluronic acid hydrogels encapsulating drug nanocrystals for long-term treatment of inflammatory arthritis. Bioeng Transl Med 2022; 7:e10245. [PMID: 35111947 PMCID: PMC8780912 DOI: 10.1002/btm2.10245] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 01/20/2023] Open
Abstract
Antiproliferative chemotherapeutic agents offer a potential effective treatment for inflammatory arthritis. However, their clinical application is limited by high systemic toxicity, low joint bioavailability as well as formulation challenges. Here, we report an intra-articular drug delivery system combining hyaluronic acid hydrogels and drug nanocrystals to achieve localized and sustained delivery of an antiproliferative chemotherapeutic agent camptothecin for long-term treatment of inflammatory arthritis. We synthesized a biocompatible, in situ-forming injectable hyaluronic acid hydrogel using a naturally occurring click chemistry: cyanobenzothiazole/cysteine reaction, which is the last step reaction in synthesizing D-luciferin in fireflies. This hydrogel was used to encapsulate camptothecin nanocrystals (size of 160-560 nm) which released free camptothecin in a sustained manner for 4 weeks. In vivo studies confirmed that the hydrogel remained in the joint over 4 weeks. By using the collagen-induced arthritis rat model, we demonstrate that the hydrogel-camptothecin formulation could alleviate arthritis severity as indicated by the joint size and interleukin-1β level in the harvested joints, as well as from histological and microcomputed tomography evaluation of joints. The hydrogel-nanocrystal formulation strategy described here offers a potential solution for intra-articular therapy for inflammatory arthritis.
Collapse
Affiliation(s)
- Yongsheng Gao
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Douglas Vogus
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Zongmin Zhao
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Wei He
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Vinu Krishnan
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Jayoung Kim
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Yujie Shi
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Apoorva Sarode
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Anvay Ukidve
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Samir Mitragotri
- School of Engineering and Applied SciencesHarvard UniversityCambridgeMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
45
|
Smith JJ, Xiao Y, Parsan N, Medwig-Kinney TN, Martinez MAQ, Moore FEQ, Palmisano NJ, Kohrman AQ, Chandhok Delos Reyes M, Adikes RC, Liu S, Bracht SA, Zhang W, Wen K, Kratsios P, Matus DQ. The SWI/SNF chromatin remodeling assemblies BAF and PBAF differentially regulate cell cycle exit and cellular invasion in vivo. PLoS Genet 2022; 18:e1009981. [PMID: 34982771 PMCID: PMC8759636 DOI: 10.1371/journal.pgen.1009981] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/14/2022] [Accepted: 12/07/2021] [Indexed: 12/15/2022] Open
Abstract
Chromatin remodelers such as the SWI/SNF complex coordinate metazoan development through broad regulation of chromatin accessibility and transcription, ensuring normal cell cycle control and cellular differentiation in a lineage-specific and temporally restricted manner. Mutations in genes encoding the structural subunits of chromatin, such as histone subunits, and chromatin regulating factors are associated with a variety of disease mechanisms including cancer metastasis, in which cancer co-opts cellular invasion programs functioning in healthy cells during development. Here we utilize Caenorhabditis elegans anchor cell (AC) invasion as an in vivo model to identify the suite of chromatin agents and chromatin regulating factors that promote cellular invasiveness. We demonstrate that the SWI/SNF ATP-dependent chromatin remodeling complex is a critical regulator of AC invasion, with pleiotropic effects on both G0 cell cycle arrest and activation of invasive machinery. Using targeted protein degradation and enhanced RNA interference (RNAi) vectors, we show that SWI/SNF contributes to AC invasion in a dose-dependent fashion, with lower levels of activity in the AC corresponding to aberrant cell cycle entry and increased loss of invasion. Our data specifically implicate the SWI/SNF BAF assembly in the regulation of the G0 cell cycle arrest in the AC, whereas the SWI/SNF PBAF assembly promotes AC invasion via cell cycle-independent mechanisms, including attachment to the basement membrane (BM) and activation of the pro-invasive fos-1/FOS gene. Together these findings demonstrate that the SWI/SNF complex is necessary for two essential components of AC invasion: arresting cell cycle progression and remodeling the BM. The work here provides valuable single-cell mechanistic insight into how the SWI/SNF assemblies differentially contribute to cellular invasion and how SWI/SNF subunit-specific disruptions may contribute to tumorigeneses and cancer metastasis. Cellular invasion is required for animal development and homeostasis. Inappropriate activation of invasion however can result in cancer metastasis. Invasion programs are orchestrated by complex gene regulatory networks (GRN) that function in a coordinated fashion to turn on and off pro-invasive genes. While the core of GRNs are DNA binding transcription factors, they require aid from chromatin remodelers to access the genome. To identify the suite of pro-invasive chromatin remodelers, we paired high resolution imaging with RNA interference to individually knockdown 269 chromatin factors, identifying the evolutionarily conserved SWItching defective/Sucrose Non-Fermenting (SWI/SNF) ATP-dependent chromatin remodeling complex as a new regulator of Caenorhabditis elegans anchor cell (AC) invasion. Using a combination of CRISPR/Cas9 genome engineering and targeted protein degradation we demonstrate that the core SWI/SNF complex functions in a dose-dependent manner to control invasion. Further, we determine that the accessory SWI/SNF complexes, BAF and PBAF, contribute to invasion via distinctive mechanisms: BAF is required to prevent inappropriate proliferation while PBAF promotes AC attachment and remodeling of the basement membrane. Together, our data provide insights into how the SWI/SNF complex, which is mutated in many human cancers, can function in a dose-dependent fashion to regulate switching from invasive to proliferative fates.
Collapse
Affiliation(s)
- Jayson J. Smith
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Yutong Xiao
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Nithin Parsan
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Taylor N. Medwig-Kinney
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Michael A. Q. Martinez
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Frances E. Q. Moore
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Nicholas J. Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Abraham Q. Kohrman
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States of America
| | - Mana Chandhok Delos Reyes
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Rebecca C. Adikes
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Biology Department, Siena College, Loudonville, New York, United States of America
| | - Simeiyun Liu
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Molecular, Cellular and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Sydney A. Bracht
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- Department of Cell Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Wan Zhang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Kailong Wen
- The Grossman Institute for Neuroscience, Quantitative Biology, and Human Behavior, University of Chicago, Chicago, Illinois, United States of America
- Department of Neurobiology, University of Chicago, Chicago, Illinois, United States of America
| | - Paschalis Kratsios
- The Grossman Institute for Neuroscience, Quantitative Biology, and Human Behavior, University of Chicago, Chicago, Illinois, United States of America
- Department of Neurobiology, University of Chicago, Chicago, Illinois, United States of America
| | - David Q. Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
46
|
Tao J, Lu Z, Su J, Qian X, Zhang Y, Xu Y, Song S, Hang X, Peng X, Chen F. ASIC1a promotes the proliferation of synovial fibroblasts via the ERK/MAPK pathway. J Transl Med 2021; 101:1353-1362. [PMID: 34282280 DOI: 10.1038/s41374-021-00636-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/21/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Synovial hyperplasia, a profound alteration in the structure of synovial tissue, is the basis for cumulative joint destruction in rheumatoid arthritis (RA). It is generally accepted that controlling synovial hyperplasia can delay the progression of RA. As one of the most intensively studied isoforms of acid-sensing ion channels (ASICs), ASIC1a contributes to various physiopathologic conditions, including RA, due to its unique property of being permeable to Ca2+. However, the role and the regulatory mechanisms of ASIC1a in synovial hyperplasia are poorly understood. Here, rats induced with adjuvant arthritis (AA) and human primary synovial fibroblasts were used in vivo and in vitro to investigate the role of ASIC1a in the proliferation of RA synovial fibroblasts (RASFs). The results show that the expression of ASIC1a was significantly increased in synovial tissues and RASFs obtained from patients with RA as well as in the synovium of rats with AA. Moreover, extracellular acidification improved the ability of RASFs colony formation and increased the expression of proliferation cell nuclear antigen (PCNA) and Ki67, which was abrogated by the specific ASIC1a inhibitor psalmotoxin-1 (PcTX-1) or ASIC1a-short hairpin RNA (ASIC1a-shRNA), suggesting that extracellular acidification promotes the proliferation of RASFs by activating ASIC1a. In addition, the activation of c-Raf and extracellular signal-regulated protein kinases (ERKs) signaling was blocked with PcTX-1 or ASIC1a-shRNA and the proliferation of RASFs was further inhibited by the ERK inhibitor (U0126), indicating that ERK/MAPK signaling contributes to the proliferation process of RASFs promoted by the activation of ASIC1a. These findings gave us an insight into the role of ASIC1a in the proliferation of RASFs, which may provide solid foundation for ASIC1a as a potential target in the treatment of RA.
Collapse
Affiliation(s)
- Jingjing Tao
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Zheng Lu
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Jingwen Su
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xuewen Qian
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yihao Zhang
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yayun Xu
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Sujing Song
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xiaoyu Hang
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Xiaoqing Peng
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Feihu Chen
- Key Laboratory of Major Autoimmune Diseases of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
47
|
Chen L, Lu Q, Chen J, Feng R, Yang C. Upregulating miR-27a-3p inhibits cell proliferation and inflammation of rheumatoid arthritis synovial fibroblasts through targeting toll-like receptor 5. Exp Ther Med 2021; 22:1227. [PMID: 34539823 PMCID: PMC8438689 DOI: 10.3892/etm.2021.10661] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis (RA) is a serious chronic inflammatory disease and synovial fibroblasts (SFs) serve a vital role in the pathogenesis and progression of RA. Current studies have demonstrated that dysregulation of microRNAs is involved in RA etiopathogenesis. The present study aimed to investigate the role of microRNA (miR)-27a-3p in RASFs, as well as its molecular mechanism. RASFs were isolated from synovial tissues from patients with RA. Expression of miR-27a-3p and toll-like receptor 5 (TLR5) was detected using reverse transcription-quantitative polymerase chain reaction and western blotting. Cell proliferation, apoptosis and inflammatory response were measured with MTT assay, flow cytometry and ELISA kits, respectively. The target binding between miR-27a-3p and TLR5 was predicted on DIANA TOOLS software, and confirmed by dual-luciferase reporter assay and Biotin-coupled miRNA pull-down assay. Expression of miR-27a-3p was downregulated and TLR5 was upregulated in synovial tissues and RASFs isolated from patients with RA. Functionally, upregulating miR-27a-3p may promote the apoptosis rate of RASFs and suppress cell proliferation and secretions of interleukin (IL)-1β, IL-6 and tumor necrosis factor-α. TLR5 was validated as a downstream target for miR-27a-3p in RASFs, and its expression was negatively regulated by miR-27a-3p. Silencing TLR5 in RASFs may exert similar effects to miR-27a-3p-overexpression; whereas, restoring TLR5 counteracted the suppression of miR-27a-3p-overexpression on RASF proliferation and inflammation, as well as the promotion on apoptosis. miR-27a-3p upregulation may suppress RA progression by inhibiting RASFs proliferation and inflammation through targeting TLR5.
Collapse
Affiliation(s)
- Lifeng Chen
- Department of Rheumatology and Immunology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Qiping Lu
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Jianhua Chen
- Department of Rheumatology and Immunology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Ruibing Feng
- Department of Orthopedics, Central People's Liberation Army Central Theater, Wuhan, Hubei 430070, P.R. China
| | - Chenxi Yang
- Department of Orthopedics, Graduate School of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| |
Collapse
|
48
|
Han Y, Wang J, Li S, Li Y, Zhang Y, Zhang R, Zhang Y, Fan H, Shi H, Pan J, Song G, Ge L, Wang L. Isopsoralen ameliorates rheumatoid arthritis by targeting MIF. Arthritis Res Ther 2021; 23:243. [PMID: 34535196 PMCID: PMC8447788 DOI: 10.1186/s13075-021-02619-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/27/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Isopsoralen (IPRN), one of the active ingredients of Psoralea corylifolia Linn, has anti-inflammatory properties. We attempted to investigate the inhibitory effects of IPRN on rheumatoid arthritis (RA) and characterize its potential mechanism. METHODS RA fibroblast-like synoviocytes (FLSs) and mice with collagen-induced arthritis (CIA) were used as in vitro and in vivo models to analyze the antiarthritic effect of IPRN. Histological analysis of the inflamed joints from mice with CIA was performed using microcomputed tomography (micro-CT) and hematoxylin-eosin (HE) staining. RNA sequencing (RNA-Seq), network pharmacology analysis, molecular docking, drug affinity responsive target stability (DARTS) assay, and cellular thermal shift assay (CETSA) were performed to evaluate the targets of IPRN. RESULTS IPRN ameliorated the inflammatory phenotype of RA FLSs by inhibiting their cytokine production, migration, invasion, and proangiogenic ability. IPRN also significantly reduced the severity of CIA in mice by decreasing paw thickness, arthritis score, bone damage, and serum inflammatory cytokine levels. A mechanistic study demonstrated that macrophage migration inhibitory factor (MIF), a key protein in the inflammatory process, was the specific target by which IPRN exerted its anti-inflammatory effects in RA FLSs. CONCLUSION Our study demonstrates the antiarthritic effect of IPRN, which suggests the therapeutic potential of IPRN in RA.
Collapse
Affiliation(s)
- Yi Han
- China Academy of Chinese Medical Sciences, Guang'anmen Hospital, Beijing, China
| | - Jinguang Wang
- Department of Orthopedics, Dezhou People's Hospital, Dezhou, Shandong, China
| | - Shufeng Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yongli Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Ruojia Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China
| | - Huancai Fan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Haojun Shi
- The Second Clinical Medical College, Henan University of Chinese Medicine, Jinan, China
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China
| | - Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China.
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Center, Key lab for Biotech-Drugs of National Health Commission, Key Lab for Rare & Uncommon Diseases of Shandong Province, Shandong First Medical University & Shandong Academy of Medical Sciences, #18877, Jingshi Road, Jinan, 250062, China.
| |
Collapse
|
49
|
Cheng X, Lu E, Fan M, Pi Z, Zheng Z, Liu S, Song F, Liu Z. A comprehensive strategy to clarify the pharmacodynamic constituents and mechanism of Wu-tou decoction based on the constituents migrating to blood and their in vivo process under pathological state. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114172. [PMID: 33932514 DOI: 10.1016/j.jep.2021.114172] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As a traditional Chinese medicine (TCM) formula, Wu-tou decoction has been used for treating rheumatoid arthritis (RA) for more than a thousand years. Identifying pharmacodynamic constituents (PCs) of WTD and exploring their in vivo process are very meaningful for promoting the modernization of TCM. However, the pathological state might change this process. AIM OF THE STUDY Hence, it is necessary and significant to compare the process in vivo of drugs both in normal and disease state and clarify their action mechanism. MATERIALS AND METHODS Taking Wu-tou decoction (WTD) as the research object, a comprehensive strategy based on liquid chromatography coupled with mass spectrometry (LC-MS) was developed to identify PCs, clarify and compare their absorption and distribution in normal and model rats, and then explore the potential mechanism of TCM. Firstly, the PCs in WTD were identified. Then, the pharmacokinetics (PK) and tissue distribution of these ingredients were studied. Finally, the constituents with the difference between normal and model rats were selected for target network pharmacological analysis to clarify the mechanism. RESULTS A total of 27 PCs of WTD were identified. The absorption and distribution of 20 PCs were successfully analyzed. In the disease state, the absorption and distribution of all these components were improved to have better treatment effects. The results of target network pharmacological analysis indicated that PTGS1, PTGS2, ABCB1, SLC6A4, CHRM2, ESR1, ESR2, CDK2, TNF and IL-6 are 10 key targets for WTD against RA. The regulatory effects of WTD on the expression of PTGS2 and TNF were further verified. Pathway enrichment analysis showed that the key mechanism of WTD against RA is to reduce inflammation and regulate the immune response. CONCLUSION These results indicated that this strategy could better understand the in vivo process and mechanism of WTD under the pathological state. Furthermore, this strategy is also appropriate for other TCM.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antirheumatic Agents/administration & dosage
- Antirheumatic Agents/chemistry
- Antirheumatic Agents/pharmacokinetics
- Antirheumatic Agents/pharmacology
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/drug therapy
- Chromatography, High Pressure Liquid
- Cyclooxygenase 2/metabolism
- Disease Models, Animal
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacokinetics
- Drugs, Chinese Herbal/pharmacology
- Glycyrrhizic Acid/blood
- Glycyrrhizic Acid/chemistry
- Inflammation/metabolism
- Lipopolysaccharides/toxicity
- Male
- Mass Spectrometry
- Medicine, Chinese Traditional
- Metabolic Networks and Pathways/drug effects
- Mice
- RAW 264.7 Cells
- Rats, Sprague-Dawley
- Tissue Distribution
- Tumor Necrosis Factor-alpha/metabolism
- Rats
Collapse
Affiliation(s)
- Xiaoxu Cheng
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, 230026, Hefei, China
| | - Enyu Lu
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, 230026, Hefei, China
| | - Meiling Fan
- Key Laboratory of Medicinal Materials, Jilin Academy of Chinese Medicine Sciences, 130021, Changchun, China
| | - Zifeng Pi
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China; Changchun Sunnytech Co.,Ltd., 130061, Changchun, China.
| | - Zhong Zheng
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China
| | - Shu Liu
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China
| | - Fengrui Song
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, 230026, Hefei, China
| | - Zhiqiang Liu
- State Key Laboratory of Electroanalytical Chemistry, National Center of Mass Spectrometry in Changchun and Jilin Provincial Key Laboratory of Chinese Medicine Chemistry and Mass Spectrometry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Changchun, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, 230026, Hefei, China.
| |
Collapse
|
50
|
Wang M, Chen Y, Bi X, Luo X, Hu Z, Liu Y, Shi X, Weng W, Mo B, Lu Y, Pan Y. LncRNA NEAT1_1 suppresses tumor-like biologic behaviors of fibroblast-like synoviocytes by targeting the miR-221-3p/uPAR axis in rheumatoid arthritis. J Leukoc Biol 2021; 111:641-653. [PMID: 34254354 DOI: 10.1002/jlb.3a0121-067rrr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Fibroblast-like synoviocytes (FLSs) are the predominant effector cells in the pathological progression of rheumatoid arthritis (RA). Therefore, elucidating the underlying molecular mechanism of the biologic behaviors in RA-FLSs will be helpful in developing the potent targets for the treatment of RA. We have previously documented that the tumor-like biologic behaviors of RA-FLSs are exacerbated by urokinase-type plasminogen activator receptor (uPAR), a specifically up-regulated receptor in RA-FLSs. Here, we investigate the further mechanism of uPAR and clarify its function in RA-FLSs. We demonstrate that miR-221-3p positively correlates to uPAR and regulates uPAR level in RA-FLSs. Simultaneously, one long noncoding RNA, nuclear paraspeckle assembly transcript 1_1 (NEAT1_1) is identified, which can predictively target miR-221-3p at three sites, indicating a strong possibility of being a competing endogenous RNA in RA-FLSs. Interestingly, NEAT1_1 and miR-221-3p can colocate in the nucleus and cytoplasm in RA-FLSs. Importantly, NEAT1_1 can act as a rheostat for the miR-221-3p/uPAR axis and the downstream JAK signaling. In line with the biologic function, NEAT1_1 negatively regulates the tumor-like characters, and cytokine secretions of RA-FLSs. Collectively, our data provide new insight into the mechanisms of NEAT1_1 in modulating RA-FLSs tumor-like behaviors. The targeting of NEAT1_1 and miR-221-3p/uPAR axis may have a promising therapeutic role in patients with RA.
Collapse
Affiliation(s)
- Manli Wang
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yixiong Chen
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuan Bi
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiqing Luo
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zuoyu Hu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Liu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Shi
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weizhen Weng
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Biyao Mo
- Division of Rheumatology, Department of Internal Medicine, Hainan General Hospital, Hainan affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yan Lu
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunfeng Pan
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|