1
|
Wang X, Tan L, Dong Y, Lin Y, Yin L, Shen S, Dou H, Hou Y. Vitamin D enhances the effect of Soufeng sanjie formula in alleviating joint inflammation in CIA mice through VDR-NOTCH3/DLL4 signaling in macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119932. [PMID: 40350051 DOI: 10.1016/j.jep.2025.119932] [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: 03/07/2025] [Revised: 04/22/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Rheumatoid arthritis (RA), defined as Bi Zheng syndrome in traditional Chinese medicine (TCM), is a chronic inflammatory and autoimmune disease that can cause substantial articular degradation and impairment. In RA, the polarization of macrophages to a proinflammatoy phenotype contributes to chronic inflammation and joint injury. Soufeng sanjie formula (SF), a traditional Chinese formula used to treat RA, consists of Scolopendra subspinipes mutilans L. Koch, Buthus martensii Karsch, Astragalus membranaceus (Fisch), and Glycine max (L.) Merr seed coats and plays a role in the regulation of macrophage polarization. Vitamin D (VD), a specific activator of the vitamin D receptor (VDR), promotes the differentiation of M2 macrophages and significantly enhances the therapeutic effects of TCM on arthritis. However, whether SF can be combined with VD to regulate macrophage polarization and alleviate RA remains unclear. AIM OF THE STUDY In this study, we examined the regulatory mechanisms and therapeutic effects of the combination of SF and VD on macrophages in RA. MATERIALS AND METHODS We used network pharmacology to analyze the targets and pathways of SF in RA. Clinical data were analyzed to confirm the expression of key targets. The RA immune landscape for cell-gene correlations was built using bioinformatics. A collagen-induced arthritis (CIA) model was established to evaluate the combined therapeutic effects of SF and VD on joint injury and inflammation. Quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting (WB), and immunofluorescence were used to assess the expression of key molecules in macrophages. Macrophage polarization was analyzed using flow cytometry. RESULTS SF acts on RA through multiple pathways, including hub genes, such as VDR, ABL1, DNMT1, and CXCR4. Immune infiltration analysis of RA showed that a reduction in the number of M2 macrophages significantly affected RA. Clinical data analysis and prognostic assessments corroborated the pivotal roles played by hub genes, such as VDR, ABL1, DNMT1, and CXCR4. Crucially, this study demonstrated a strong association between the key hub gene VDR and M2 macrophages. In vivo and in vitro models of RA showed that VD significantly improved the efficacy of SF against arthritis in CIA mice and stimulated macrophage polarization toward the M2 phenotype via modulation of the NOTCH3/DLL4 pathway. Furthermore, siRNA-mediated interference with the expression of VDR confirmed that the downregulation of VDR significantly activated the NOTCH3/DLL4 signaling pathway and blocked the regulation of SF combined with VD on the polarization of macrophages toward the M2 phenotype. CONCLUSIONS Our results demonstrate that the combination of SF and VD markedly improves the therapeutic effect of SF on RA via M2 macrophage differentiation through the VDR-NOTCH3/DLL4 signaling pathway. The VD-enhancing effect of SF in improving RA symptoms offers a new strategy and theoretical foundation for the clinical treatment of the condition.
Collapse
Affiliation(s)
- Xiuzhu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Liping Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Yue Dong
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Yan Lin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Lijie Yin
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Sunan Shen
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China.
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China.
| |
Collapse
|
2
|
Tang J, Li S, Wang Y, Yuan M, Wan Y, Liang X, Guo L, Guo Y. Study of Folate-Modified Carboxymethyl Chitosan-Sinomenine-Curcumin Nanopolymer for Targeted Treatment of Rheumatoid Arthritis. Biomacromolecules 2025. [PMID: 40324022 DOI: 10.1021/acs.biomac.4c01701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Sinomenine hydrochloride (SH) has been clinically utilized for many years to treat rheumatoid arthritis (RA) in both oral and injectable forms. However, its low bioavailability, poor targeting, high dosage requirements, and side effects, present significant challenges. This study developed folic acid-carboxymethyl chitosan-modified sinomenine-curcumin nanopolymers (named SCNP) for the targeted treatment of RA, to reduce dosage and side effects. The design of SCNP employs folic acid (FA) as a targeting moiety, facilitating specific binding to the folate receptor (FR) on the surface of macrophages and enabling internalization into activated macrophages via endocytosis, thereby achieving targeted delivery to sites of inflammation. In a rat and cell model of RA, SCNP was found to decrease reactive oxygen species (ROS) and pro-inflammatory factors while increasing the anti-inflammatory factor IL-10 through the NF-κB/NLRP3 pathway. These findings indicate that SCNP has the potential to lower drug dosage, enhance therapeutic efficacy, and minimize side effects such as diarrhea and rash, thereby highlighting its promise as an inflammation-targeting nanopolymer.
Collapse
Affiliation(s)
- Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xue Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
3
|
Zhang L, Wu G, Yao J, Wang D, Gao F, Qian Z. Hyaluronic acid-modified PtPdCo-CQ nanocatalyst with triple enzyme-like activities regulates macrophage polarization and autophagy levels for the treatment of rheumatoid arthritis. Int J Biol Macromol 2025; 309:143143. [PMID: 40233904 DOI: 10.1016/j.ijbiomac.2025.143143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/30/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial inflammation and imbalanced macrophage polarization. Pro-inflammatory M1 macrophages exacerbate joint damage through excessive production of reactive oxygen species (ROS), while anti-inflammatory M2 macrophages are prone to ferroptosis, limiting the long-term efficacy of existing nanozyme therapies. This study aimed to develop a novel nanocatalyst combining efficient ROS scavenging and M2 macrophage protection to synergistically regulate macrophage polarization and autophagy levels for sustained RA remission. We designed a hyaluronic acid-modified PtPdCo-CQ nanocatalyst (HPPCQ) with triple enzyme-like activities-superoxide dismutase (SOD), catalase (CAT), and peroxidase (POD). In vitro experiments demonstrated that HPPCQ activated the Nrf2/HO-1 antioxidant pathway by scavenging ROS, promoted M1-to-M2 phenotypic repolarization, and protected M2 macrophages from autophagy-dependent ferroptosis via controlled release of chloroquine (CQ). In a collagen-induced arthritis (CIA) mouse model, HPPCQ targeted inflamed joints, significantly reducing clinical scores, synovial hyperplasia, and pro-inflammatory cytokine levels (TNF-α, IL-1β). Histological analysis revealed markedly alleviated cartilage destruction and inflammatory infiltration in HPPCQ-treated mice. By integrating ROS scavenging, macrophage reprogramming, and ferroptosis inhibition, this work provides a novel therapeutic strategy with enhanced efficacy and durability for RA treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China; Department of Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Emergency, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Guoquan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, China
| | - Junwei Yao
- Department of Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China
| | - Dajun Wang
- Department of Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Provincial Institute of Health Emergency, Xuzhou Medical University, Xuzhou, China
| | - Fenglei Gao
- Department of Emergency Center, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, China.
| | - Zhonglai Qian
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Vargas-Ángeles CA, Trujillo-Cirilo L, Sierra-Mondragón E, Rangel-Corona R, Weiss-Steider B. Cationic liposomes encapsulating IL-2 selectively induce apoptosis and significantly reduce the secretion of cytokines on M1-murine polarized macrophages. Cytokine 2025; 189:156903. [PMID: 40023103 DOI: 10.1016/j.cyto.2025.156903] [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: 10/31/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Inflammatory diseases pose a global challenge due to the critical role of macrophages in their pathogenesis. This study evaluated the effects of cationic liposomes encapsulating IL-2 (LIP-IL-2) on murine peritoneal macrophages (MP) polarized towards M1 and M2 phenotypes. M1 MP (CD86+), which overexpressed IL-2Rα and CD14 receptors, underwent apoptosis following LIP-IL-2 treatment, whereas M2 MP (CD206+) were unaffected. Furthermore, LIP-IL-2 significantly reduced the secretion of pro-inflammatory cytokines, including IL-6, TNF-α, IFN-γ, and IL-12, exclusively in M1 macrophages. These findings suggest that LIP-IL-2 could serve as a promising therapeutic tool for chronic inflammatory diseases by selectively inducing apoptosis in pro-inflammatory M1 macrophages without affecting M2 ones, opening avenues for targeted therapies in diseases where chronic macrophage-mediated inflammation is a key factor.
Collapse
Affiliation(s)
- C A Vargas-Ángeles
- Laboratory of Cellular Oncology, Cell Differentiation and Cancer Unit, L-4 PB, FES Zaragoza, National Autonomous University of Mexico, Av. Guelatao No 66 Col. Ejercito de Oriente, Iztapalapa, CP 09230, México City, Mexico; Graduate Program in Biological Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - L Trujillo-Cirilo
- Laboratory of Cellular Oncology, Cell Differentiation and Cancer Unit, L-4 PB, FES Zaragoza, National Autonomous University of Mexico, Av. Guelatao No 66 Col. Ejercito de Oriente, Iztapalapa, CP 09230, México City, Mexico
| | - E Sierra-Mondragón
- Laboratory of Cellular Oncology, Cell Differentiation and Cancer Unit, L-4 PB, FES Zaragoza, National Autonomous University of Mexico, Av. Guelatao No 66 Col. Ejercito de Oriente, Iztapalapa, CP 09230, México City, Mexico
| | - R Rangel-Corona
- Laboratory of Cellular Oncology, Cell Differentiation and Cancer Unit, L-4 PB, FES Zaragoza, National Autonomous University of Mexico, Av. Guelatao No 66 Col. Ejercito de Oriente, Iztapalapa, CP 09230, México City, Mexico.
| | - B Weiss-Steider
- Laboratory of Cellular Oncology, Cell Differentiation and Cancer Unit, L-4 PB, FES Zaragoza, National Autonomous University of Mexico, Av. Guelatao No 66 Col. Ejercito de Oriente, Iztapalapa, CP 09230, México City, Mexico
| |
Collapse
|
5
|
Zhang Z, Liu Y, Liang X, Wang Q, Xu M, Yang X, Tang J, He X, He Y, Zhang D, Li C. Advances in nanodelivery systems based on apoptosis strategies for enhanced rheumatoid arthritis therapy. Acta Biomater 2025; 197:87-103. [PMID: 40154765 DOI: 10.1016/j.actbio.2025.03.043] [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: 11/26/2024] [Revised: 03/11/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disorder primarily characterized by persistent synovial inflammation and progressive bone erosion. The pathogenesis of RA involves a complex cascade of cellular and molecular events, including sustained hyperactivation of macrophages, excessive recruitment and activation of neutrophils, pathological proliferation and invasion of fibroblast-like synoviocytes (FLS), and dysregulated differentiation and function of osteoclasts (OCs). The inflammatory factors secreted by these dysregulated cells significantly disrupt the joint microenvironment through multiple pathological mechanisms, primarily by promoting synovial inflammation, cartilage matrix degradation, osteoclast-mediated bone erosion, and pathological angiogenesis. Therapeutic strategies targeting the induction of apoptosis in these malignant cells have demonstrated considerable potential in preclinical studies, offering a promising approach to enhance treatment outcomes by simultaneously reducing inflammatory cytokine production and inhibiting pathogenic cell proliferation. However, conventional therapeutic drugs are limited in clinical applications because of their high toxicity and side effects. Inflammation induces morphological and functional changes in cells within the rheumatoid arthritis microenvironment (RAM), particularly the overexpression of specific receptors on cell membranes. This phenomenon has driven the development of ligand-modified targeted nanodelivery systems (NDSs), which can specifically target and induce apoptosis in specific cell types, thereby enhancing therapeutic efficacy. This paper comprehensively reviews the research progress of targeted NDSs based on apoptosis strategies for RA therapy, with a detailed discussion of their advantages in inducing apoptosis in various disease-associated cells. Furthermore, the potential of combining apoptosis of multiple cell types for RA treatment is explored. This review is expected to improve insights into the apoptosis of malignant cells to enhance RA therapy. STATEMENT OF SIGNIFICANCE: This review highlights recent advances in nanodelivery systems (NDSs) based on apoptotic strategies for enhanced rheumatoid arthritis (RA) therapy. Unlike conventional NDSs, these optimized systems specifically induce apoptosis in malignant cells within the RA microenvironment by integrating multiple therapeutic strategies. By summarizing the latest research, our work demonstrates the potential of these NDSs to suppress inflammatory responses and prevent bone destruction through targeted elimination of malignant cells, offering a novel direction for RA treatment. This review is significant as it provides a comprehensive overview for researchers and clinicians, facilitating the development of more effective therapeutic approaches for RA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Zongquan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yilin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qian Wang
- Classical teaching and Research Department, College of Integrated Chinese and Western medicine, Affiliated TCM Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Maochang Xu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xi Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jun Tang
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xinghui He
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Yufeng He
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Dan Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Green Pharmaceutical Technology Key Laboratory of Luzhou, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
6
|
Wang R, Ji C, Chen J, Zhang X, Hu Q, Liu C. Research advances in the treatment of arthritis from natural products (2014-present). Chin J Nat Med 2025; 23:529-540. [PMID: 40383610 DOI: 10.1016/s1875-5364(25)60862-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 05/20/2025]
Abstract
Arthritis, encompassing osteoarthritis (OA), rheumatoid arthritis (RA), and gouty arthritis (GA), is a prevalent inflammatory disease that significantly impacts quality of life. Natural products (NPs), derived from animals, plants, marine organisms, and microorganisms, have demonstrated beneficial effects in arthritis treatment both domestically and internationally. These natural compounds offer advantages in drug discovery due to their skeletal diversity, structural complexity, and multi-effect, multi-target, and low-toxicity properties compared to conventional small-molecule medicines. However, unclear mechanisms have hindered the development and clinical application of NPs. This review summarizes recent experimental studies from the past decade on natural medicine for arthritis treatment, emphasizing key NPs with therapeutic effects on OA, RA, and GA. It examines the effects and molecular mechanisms of NPs acting on different cells to treat arthritis. Furthermore, this review provides insights into the future prospects of NP research in this field, which is crucial for advancing NP-based arthritis treatments.
Collapse
Affiliation(s)
- Ruilin Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Cen Ji
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiayao Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaohan Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qinghua Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Chunxiao Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China; School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
7
|
Li L, Guo H, Zhang W, Xiang X, Chi J, Zhang M, Chen J, Wang Z, Dai L. Biomarker identification for rheumatoid arthritis with inadequate response to DMARD and TNF therapies using multidimensional analyses. Immunobiology 2025; 230:152901. [PMID: 40288069 DOI: 10.1016/j.imbio.2025.152901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE Rheumatoid arthritis (RA) is an immune system disorder disease accompanied with severe joint damage. However, the molecular mechanism of RA with insensitive to medicine remains insufficient. Thus, this study aims to identify the biomarkers of RA patients with inadequate responses (IR) toward disease-modifying antirheumatic drug (DMARD) and antitumor necrosis factor (TNF) therapies, using multidimensional analyses. METHODS Gene expression data GSE45291 originating from clinics were downloaded from the Gene Expression Omnibus public database (GEO). Differentially expressed genes (DEGs) closely associated with DMARD&TNF-IR RA were identified using the Limma R package. Weighted gene co-expression network analysis (WGCNA) was carried out to identify critical genes. The CIBERSORT algorithm and single sample Gene Set Enrichment Analysis (ssGSEA) were employed for immune infiltration analysis and functional enrichment analysis, respectively. Lastly, mRNA expression levels of the identified hub genes in inflammatory conditions of collagen-induced arthritis (CIA) rats and lipopolysaccharide (LPS)-induced RAW264.7 cells were further observed using RT-qPCR. RESULTS In this work, a total of 17 genes were identified as hub genes. Of these, the expression levels of UHMK1, ELK4, APOC2, and SFT2D1 were significantly lowered in inflammatory conditions. GSEA indicated B cells with the immune-related genes play an essential role in the course of DMARD&TNF-IR RA. Notable differences in immune cell proportions (activated. Dendritic. cell, CD56 bright. Natural. killer. Cell, gamma. Delta. T. cell, MDSC, macrophage) were observed between normal and disease groups, suggesting immune involvement. CONCLUSION The findings of this study provide additional understanding of the detection of DMARD&TNF-IR RA.
Collapse
Affiliation(s)
- Leyuan Li
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Hui Guo
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China; Engineering Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, HUCM, Zhengzhou 450015, China
| | - Weijin Zhang
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Xi Xiang
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Jun Chi
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China; Engineering Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, HUCM, Zhengzhou 450015, China
| | - Mengmeng Zhang
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China
| | - Jiali Chen
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhimin Wang
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Liping Dai
- Henan University of Chinese Medicine (HUCM), Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou 450046, China; Engineering Center for Comprehensive Development and Utilization of Authentic Medicinal Materials in Henan Province, HUCM, Zhengzhou 450015, China.
| |
Collapse
|
8
|
Barbalace MC, Talotta R, Rapisarda F, D’Amico V, Laganà M, Malaguti M, Campennì A, Cannavò S, Hrelia S, Ruggeri RM. Unlocking the Power of the Mediterranean Diet: Two in One-Dual Benefits for Rheumatic and Thyroid Autoimmune Diseases. Nutrients 2025; 17:1383. [PMID: 40284245 PMCID: PMC12030468 DOI: 10.3390/nu17081383] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
In recent years, autoimmune diseases are becoming more and more prevalent worldwide, with this rapid rise being influenced by environmental factors linked to lifestyle changes in modern societies. In this context, the role of diet has been the topic of extensive research as evidence has mounted that particular dietary patterns may contribute to or modulate autoimmunity. The present review specifically focuses on the Mediterranean diet (MD) as a whole dietary pattern, and on its peculiar components, such as n-3 polyunsaturated fatty acids (PUFAs), polyphenols and fiber. We explored their potential benefits in a spectrum of both systemic and organ-specific autoimmune disorders, including rheumatic diseases (like rheumatic arthritis and systemic lupus erythematosus), and thyroid diseases (like Hashimoto's thyroiditis), since they often occur in the same individuals. Here, we offer a comprehensive review about the influence of dietary factors on these autoimmune diseases and potential translation into therapeutic interventions, as an adjuvant therapeutic approach to improve autoimmunity-related outcomes.
Collapse
Affiliation(s)
- Maria Cristina Barbalace
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.M.); (S.H.)
| | - Rossella Talotta
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, 98124 Messina, Italy; (R.T.); (F.R.); (V.D.)
| | - Federica Rapisarda
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, 98124 Messina, Italy; (R.T.); (F.R.); (V.D.)
| | - Valeria D’Amico
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University Hospital “G. Martino”, 98124 Messina, Italy; (R.T.); (F.R.); (V.D.)
| | - Martina Laganà
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV “G. Barresi”, University of Messina, 98125 Messina, Italy; (M.L.); (S.C.); (R.M.R.)
| | - Marco Malaguti
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.M.); (S.H.)
| | - Alfredo Campennì
- Unit of Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy;
| | - Salvatore Cannavò
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV “G. Barresi”, University of Messina, 98125 Messina, Italy; (M.L.); (S.C.); (R.M.R.)
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47921 Rimini, Italy; (M.M.); (S.H.)
| | - Rosaria Maddalena Ruggeri
- Endocrinology Unit, Department of Human Pathology of Adulthood and Childhood DETEV “G. Barresi”, University of Messina, 98125 Messina, Italy; (M.L.); (S.C.); (R.M.R.)
| |
Collapse
|
9
|
Cai W, Wu A, Lin Z, Cao W, Pathak JL, Jaspers RT, Li R, Li X, Zheng K, Lin Y, Zhou N, Zhang X, Zhu Y, Zhang Q. S-propargyl-cysteine attenuates temporomandibular joint osteoarthritis by regulating macrophage polarization via Inhibition of JAK/STAT signaling. Mol Med 2025; 31:128. [PMID: 40197110 PMCID: PMC11974036 DOI: 10.1186/s10020-025-01186-6] [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: 12/18/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Temporomandibular joint osteoarthritis (TMJ-OA) is a disease characterized by cartilage degradation and synovial inflammation, with limited effective treatment currently. Synovial macrophage polarization is pivotal in TMJ-OA progression, making it a promising therapeutic aspect. This study investigated the effects of S-propargyl-cysteine (SPRC), an endogenous H2S donor, on macrophage polarization and its therapeutic potential in alleviating TMJ-OA. METHODS A MIA-induced TMJ-OA rat model and LPS-stimulated RAW264.7 macrophages were employed to evaluate the effects of SPRC in vivo and in vitro. TMJ bone and cartilage were analyzed via micro-CT and histological methods, while macrophage polarization markers expression were assessed via RT-qPCR, western blot, and immunofluorescence. RNA sequencing was performed on macrophages, and the JAK2/STAT3 signaling pathway was validated using the JAK2-specific inhibitor AG490. The direct effects of SPRC on rat primary condylar chondrocytes were examined by evaluating ECM synthesis and degradation. Co-culture experiments further assessed macrophage-chondrocyte interactions. RESULTS SPRC significantly alleviated cartilage and bone damage in the TMJ-OA rat model, as demonstrated by improved bone volume and cartilage structure. SPRC reduced pro-inflammatory M1 macrophage infiltration and enhanced anti-inflammatory M2 macrophage polarization. SPRC effectively inhibited the JAK2/STAT3, leading to reduction of inflammatory markers, including TNF-α, IL-6, and iNOS. Co-culture experiments revealed that SPRC-treated macrophage-conditioned medium improved chondrocyte metabolic activity and restored ECM integrity. CONCLUSIONS SPRC-modulated macrophage polarization alleviates TMJ-OA via JAK/STAT downregulation, thereby reducing synovial inflammation and cartilage degradation. These findings position SPRC as a promising therapeutic candidate for TMJ-OA and provide insights into novel strategies targeting macrophage polarization and synovium-cartilage crosstalk.
Collapse
Affiliation(s)
- Wenyi Cai
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Antong Wu
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Zhongxiao Lin
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wei Cao
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Janak L Pathak
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Richard T Jaspers
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Rui Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Xin Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Kaihan Zheng
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Yufu Lin
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Na Zhou
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
| | - Xin Zhang
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
| | - Yizhun Zhu
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China.
| | - Qingbin Zhang
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China.
| |
Collapse
|
10
|
Jafarzadeh S, Nemati M, Zandvakili R, Jafarzadeh A. Modulation of M1 and M2 macrophage polarization by metformin: Implications for inflammatory diseases and malignant tumors. Int Immunopharmacol 2025; 151:114345. [PMID: 40024215 DOI: 10.1016/j.intimp.2025.114345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/04/2025]
Abstract
Macrophages perform an essential role in the body's defense mechanisms and tissue homeostasis. These cells exhibit plasticity and are categorized into two phenotypes, including classically activated/M1 pro-inflammatory and alternatively activated/M2 anti-inflammatory phenotypes. Functional deviation in macrophage polarization occurs in different pathological conditions that need correction. In addition to antidiabetic impacts, metformin also possesses multiple biological activities, including immunomodulatory, anti-inflammatory, anti-tumorigenic, anti-aging, cardioprotective, hepatoprotective, and tissue-regenerative properties. Metformin can influence the polarization of macrophages toward M1 and M2 phenotypes. The ability of metformin to support M2 polarization and suppress M1 polarization could enhance its anti-inflammatory properties and potentiate its protective effects in conditions such as chronic inflammatory diseases, atherosclerosis, and obesity. However, in metformin-treated tumors, the proportion of M2 macrophages is decreased, while the frequency ratio of M1 macrophages is increased, indicating that metformin can modulate macrophage polarization from a pro-tumoral M2 state to an anti-tumoral M1 phenotype in malignancies. Metformin affects macrophage polarization through AMPK-dependent and independent pathways involving factors, such as NF-κB, mTOR, ATF, AKT/AS160, SIRT1, STAT3, HO-1, PGC-1α/PPAR-γ, and NLRP3 inflammasome. By modulating cellular metabolism and apoptosis, metformin can also influence macrophage polarization. This review provides comprehensive evidence regarding metformin's effects on macrophage polarization and the underlying mechanisms. The polarization-inducing capabilities of metformin may provide significant therapeutic applications in various inflammatory diseases and malignant tumors.
Collapse
Affiliation(s)
- Sara Jafarzadeh
- Student Research Committee, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran; Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Raziyeh Zandvakili
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
11
|
Ye X, Ren D, Chen Q, Shen J, Wang B, Wu S, Zhang H. Resolution of inflammation during rheumatoid arthritis. Front Cell Dev Biol 2025; 13:1556359. [PMID: 40206402 PMCID: PMC11979130 DOI: 10.3389/fcell.2025.1556359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes synovial joint inflammation as well as bone destruction and erosion, typically characterized by joint pain, swelling, and stiffness, with complications and persistent pain after remission posing a significant health burden for RA patients. The etiology of RA has not yet been fully elucidated, but a large number of studies have shown that the initiation of inflammation in RA is closely related to T-cell activation, the production of a variety of pro-inflammatory cytokines, macrophage M1/M2 imbalance, homeostatic imbalance of the intestinal flora, fibroblast-like synoviocytes (FLSs) and synovial tissue macrophages (STMs) in the synovial lumen of joints that exhibit an aggressive phenotype. While the resolution of RA is less discussed, therefore, we provided a systematic review of the relevant remission mechanisms including blocking T cell activation, regulating macrophage polarization status, modulating the signaling pathway of FLSs, modulating the subpopulation of STMs, and inhibiting the relevant inflammatory factors, as well as the probable causes of persistent arthritis pain after the remission of RA and its pain management methods. Achieving resolution in RA is crucial for improving the quality of life and long-term prognosis of patients. Thus, understanding these mechanisms provide novel potential for further drug development and treatment of RA.
Collapse
Affiliation(s)
- Xiaoou Ye
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Dan Ren
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Qingyuan Chen
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Jiquan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Bo Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Lishui University, Lishui, China
- Wenzhou Medical University Affiliated Lishui Hospital, Lishui, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| |
Collapse
|
12
|
Jiang Y, Wang S, Zhu W, Liu X, Yang Y, Huo L, Ye J, Ma Y, Zhou Y, Yang Z, Mao J, Wang X. Lysyl Oxidase-Like 1 (LOXL1) Up-Regulation in Chondrocytes Promotes M1 Macrophage Activation in Osteoarthritis via NF-κB and STAT3 Signaling. Immunotargets Ther 2025; 14:259-278. [PMID: 40161479 PMCID: PMC11951931 DOI: 10.2147/itt.s512768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/16/2025] [Indexed: 04/02/2025] Open
Abstract
Purpose Osteoarthritis (OA) constitutes a widespread degenerative joint disease predominantly affecting the elderly, leading to disability. There is still a lack of biomarkers for OA, so it cannot be intervened in time. Methods OA biomarkers were identified from human cartilage datasets using LASSO and SVM-RFE, followed by ROC analysis. LOXL1 was prioritized for further research due to its high expression in OA cartilage and robust predictive performance. Anterior cruciate ligament transection (ACLT) surgery-induced OA rats were used to explore the correlation between LOXL1 and inflammatory factors and macrophages. Macrophage markers and cytokine secretion were detected from macrophages treated with LOXL1, or co-cultured with chondrocytes after LOXL1 siRNA silencing. Results Five hub biomarkers with OA-specific expression were identified. Elevated LOXL1 correlated with IL-6 and IL-8 in patients and increased M1 macrophages in OA rats. LOXL1-stimulated macrophages upregulated CD86 and inflammatory cytokines. Silencing LOXL1 in chondrocytes reduced CD86, inflammatory cytokines, and NF-κB p65 and p-STAT3 expression in co-cultured macrophages, mitigating MMP13 and chondrocyte apoptosis. STAT3 and NF-κB signal inhibition reduces p-STAT3, p-p65, CD86, IL-6 and IL-1β expression in LOXL1-stimulated macrophages. Conclusion This study underscores the pivotal role of LOXL1 in activating M1 macrophages through NF-κB and STAT3 signaling, thereby promoting pro-inflammatory cytokine secretion and contributing to OA pathogenesis. LOXL1 holds promise as a potential marker for early diagnosis of OA inflammation and as a novel therapeutic target.
Collapse
Affiliation(s)
- Yuyun Jiang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Shang Wang
- Tzu Chi International College of Traditional Chinese Medicine, Vancouver, BC, Canada
| | - Wei Zhu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
- Department of Sports Medicine, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Xi Liu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Yanwei Yang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Liyue Huo
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jixian Ye
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Yongbin Ma
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
- Department of Central Laboratory, Jintan Hospital, Jiangsu University, Jintan, 213200, People’s Republic of China
| | - Yuepeng Zhou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhe Yang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jiahui Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
- Department of Nuclear Medicine, Institute of Digestive Diseases, and Institute of Endocrinology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
13
|
Piscitelli E, Abeni E, Balbino C, Angeli E, Cocola C, Pelucchi P, Palizban M, Diaspro A, Götte M, Zucchi I, Reinbold RA. Glycosylation Regulation by TMEM230 in Aging and Autoimmunity. Int J Mol Sci 2025; 26:2412. [PMID: 40141059 PMCID: PMC11942208 DOI: 10.3390/ijms26062412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Aging is often a choice between developing cancer or autoimmune disorders, often due in part to loss of self-tolerance or loss of immunological recognition of rogue-acting tumor cells. Self-tolerance and cell recognition by the immune system are processes very much dependent on the specific signatures of glycans and glycosylated factors present on the cell plasma membrane or in the stromal components of tissue. Glycosylated factors are generated in nearly innumerable variations in nature, allowing for the immensely diverse role of these factors in aging and flexibility necessary for cellular interactions in tissue functionality. In previous studies, we showed that differential expression of TMEM230, an endoplasmic reticulum (ER) protein was associated with specific signatures of enzymes regulating glycan synthesis and processing and glycosylation in rheumatoid arthritis synovial tissue using single-cell transcript sequencing. In this current study, we characterize the genes and pathways co-modulated in all cell types of the synovial tissue with the enzymes regulating glycan synthesis and processing, as well as glycosylation. Genes and biological and molecular pathways associated with hallmarks of aging were in mitochondria-dependent oxidative phosphorylation and reactive oxygen species synthesis, ER-dependent stress and unfolded protein response, DNA repair (UV response and P53 signaling pathways), and senescence, glycolysis and apoptosis regulation through PI3K-AKT-mTOR signaling have been shown to play important roles in aging or neurodegeneration (such as Parkinson's and Alzheimer's disease). We propose that the downregulation of TMEM230 and RNASET2 may represent a paradigm for the study of age-dependent autoimmune disorders due to their role in regulating glycosylation, unfolded protein response, and PI3K-AKT-mTOR signaling.
Collapse
Affiliation(s)
- Eleonora Piscitelli
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (E.P.); (E.A.); (C.C.); (P.P.)
| | - Edoardo Abeni
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (E.P.); (E.A.); (C.C.); (P.P.)
| | | | - Elena Angeli
- Department of Physics, University of Genoa, 16146 Genoa, Italy; (E.A.); (A.D.)
| | - Cinzia Cocola
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (E.P.); (E.A.); (C.C.); (P.P.)
| | - Paride Pelucchi
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (E.P.); (E.A.); (C.C.); (P.P.)
| | - Mira Palizban
- Department of Gynecology and Obstetrics, University Hospital of Münster, 48149 Münster, Germany (M.G.)
| | - Alberto Diaspro
- Department of Physics, University of Genoa, 16146 Genoa, Italy; (E.A.); (A.D.)
- Nanoscopy, Istituto Italiano Tecnologia, 16152 Genoa, Italy
| | - Martin Götte
- Department of Gynecology and Obstetrics, University Hospital of Münster, 48149 Münster, Germany (M.G.)
| | - Ileana Zucchi
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (E.P.); (E.A.); (C.C.); (P.P.)
- Associazione Fondazione Renato Dulbecco, Via Fantoli 16/15, 20138 Milan, Italy
| | - Rolland A. Reinbold
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Italy; (E.P.); (E.A.); (C.C.); (P.P.)
- Associazione Fondazione Renato Dulbecco, Via Fantoli 16/15, 20138 Milan, Italy
| |
Collapse
|
14
|
Chen J, Xu D, Chen B, Jian S, Chen T, Wang W, Ma Y, Zhang J, Li K, Cai W, Xiao M. Senescent macrophages trigger a pro-inflammatory program and promote the progression of rheumatoid arthritis. Int Immunopharmacol 2025; 149:114164. [PMID: 39904033 DOI: 10.1016/j.intimp.2025.114164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/27/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
This study explores the complex mechanisms between Rheumatoid Arthritis (RA) and cellular senescence, with a focus on the role of four key genes (MMP1, CCL7, CXCL1, HK3) identified through transcriptome analysis in the GEO database. These genes are closely related to IL-17 signalling and the pathogenesis of RA. In a macrophage senescence model induced by hydrogen peroxide (H2O2) and bleomycin (BLM), quantitative real-time PCR (qRT-PCR) and Western blot confirmed the significant upregulation of these genes and an increase in the secretion of the cytokine IL-17, which promotes an inflammatory environment for the polarization of macrophages to M1. When co-cultured with mouse synovial fibroblasts (MSF), MSF showed enhanced vitality and increased invasiveness, indicating a key role for these genes in the progression of RA. Additionally, HK3, less reported in RA, when its expression is knocking down, lactate secretion and lactylation modification at lysine 14 of histone H3 in macrophages were reduced, leading to a change in macrophage polarity. The study concludes that the altered polarity of senescent macrophages drives the proliferation and invasion of MSF, significantly promoting the development of RA and providing insights into the pathophysiology of RA.
Collapse
Affiliation(s)
- Jiayao Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Dahua Xu
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou 571199 Hainan, China; Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou 571199 Hainan, China
| | - Bocen Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Shaoqin Jian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Tong Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Wenguang Wang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199 Hainan, China
| | - Yingjie Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199 Hainan, China
| | - Jing Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China
| | - Kongning Li
- College of Biomedical Information and Engineering, Hainan Medical University, Haikou 571199 Hainan, China; Hainan Engineering Research Center for Health Big Data, Hainan Medical University, Haikou 571199 Hainan, China.
| | - Wangwei Cai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China.
| | - Man Xiao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou 571199 Hainan, China.
| |
Collapse
|
15
|
Liu L, Li M, Zhang C, Zhong Y, Liao B, Feng J, Deng L. Macrophage metabolic reprogramming: A trigger for cardiac damage in autoimmune diseases. Autoimmun Rev 2025; 24:103733. [PMID: 39716498 DOI: 10.1016/j.autrev.2024.103733] [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: 10/09/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024]
Abstract
Macrophage metabolic reprogramming has a central role in the progression of autoimmune and auto-inflammatory diseases. The heart is a major target organ in many autoimmune conditions and can sustain functional and structural impairments, potentially leading to irreversible cardiac damage. There is mounting clinical evidence pointing to a link between autoimmune disease and cardiac damage. However, this association remains poorly understood, and numerous patients do not receive appropriate preventive measures, which poses serious cardiovascular risks and significantly impacts their quality of life. This review discusses the relationship between macrophage metabolic reprogramming and cardiac damage in patients with autoimmune diseases and the role of adaptive immunity in macrophage reprogramming. It also provides an overview of the immunosuppressive therapies used at present. Exploiting the properties of macrophage reprogramming could lead to development of novel treatments for patients with autoimmune-related cardiac damage.
Collapse
Affiliation(s)
- Lin Liu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Minghao Li
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Chunyu Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China
| | - Bin Liao
- Department of Cardiovascular Surgery, The Affiliated Hospital of Southwest Medical University, Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Luzhou, China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China.
| | - Li Deng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Stem Cell Immunity and Regeneration Key Laboratory of Luzhou, Luzhou, China; Department of Rheumatology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
16
|
Zhang Z, Yuan D, Jin X, Chang W, Zhang Y, Xie W. Asperosaponin VI suppresses ferroptosis in chondrocytes and ameliorates osteoarthritis by modulating the Nrf2/GPX4/HO-1 signaling pathway. Front Pharmacol 2025; 16:1539092. [PMID: 40093317 PMCID: PMC11906723 DOI: 10.3389/fphar.2025.1539092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Background Asperosaponin VI (AVI) is a naturally occurring monosaccharide derived from Dipsacus asperoides renowned for its anti-inflammatory and bone-protective properties. Objective To elucidate the specific mechanism through which AVI affects chondrocytes in osteoarthritis (OA). Methods For the in vitro experiments, primary chondrocytes were to elucidate the molecular mechanisms underlying the action of AVI.For the in vivo experiments, rat OA models were established using a modified Hulth method. The severity of knee osteoarthritis was evaluated 8 weeks post-surgery. Micro-CT imaging, hematoxylin-eosin staining, and Safranin O-fast green staining were used to assess degeneration in rat knee joints. Immunohistochemistry techniques were conducted to measure the levels of collagen II, MMP13, Nrf2, GPX4, ACSL4, and HO-1 within cartilage tissues. ELISA assays were performed to measure those of TNF-α, IL -6, and PGE2 in serum samples. Results AVI alleviated chondrocyte apoptosis and extracellular matrix degradation in rat OA induced by IL-1β. It attenuated the levels of TNF-α, IL-6, and PGE2 while reducing those of Fe2+ and malondialdehyde (MDA). AVI upregulated the expression of Nrf2, HO-1, and GPX4 while downregulating that of ACSL4. Mechanistic studies revealed that ML385-induced inhibition of the Nrf2 signaling pathway reversed the increase in GPX4 and ACSL4 expression and increased Fe2+ and MDA levels; treatment with erastin, a ferroptosis inducer, produced comparable results. In vivo experiments demonstrated that AVI improved the bone volume/tissue volume and trabecular separation values in OA rats; reversed the Osteoarthritis Research Society International score; upregulated Nrf2, HO-1, and GPX4 expression; downregulated ACSL4 and MMP13 expression, and decreased the serum levels of TNF-α, IL-6, and PGE2. Conclusion Our findings suggest that AVI is a promising therapeutic agent for OA. It exerted its protective effect by regulating the Nrf2/GPX4/HO-1 signaling axis to inhibit cartilage cell ferroptosis and improve osteoarthritis.
Collapse
Affiliation(s)
- Zhimeng Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Daotong Yuan
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ximin Jin
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenjie Chang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yongkui Zhang
- Department of Orthopedic Surgery, Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenpeng Xie
- Department of Orthopedic Surgery, Affliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
17
|
Wu Z, Zhan W, Wu L, Yu L, Xie X, Yu F, Kong W, Bi S, Liu S, Yin G, Zhou J. The Roles of Forkhead Box O3a (FOXO3a) in Bone and Cartilage Diseases - A Narrative Review. Drug Des Devel Ther 2025; 19:1357-1375. [PMID: 40034405 PMCID: PMC11874768 DOI: 10.2147/dddt.s494841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
Bone and cartilage diseases are significantly associated with musculoskeletal disability. However, no effective drugs are available to cure them. FOXO3a, a member of the FOXO family, has been implicated in cell proliferation, ROS detoxification, autophagy, and apoptosis. The biological functions of FOXO3a can be modulated by post-translational modifications (PTMs), such as phosphorylation and acetylation. Several signaling pathways, such as MAPK, NF-κB, PI3K/AKT, and AMPK/Sirt1 pathways, have been implicated in the development of bone and cartilage diseases by mediating the expression of FOXO3a. In particular, FOXO3a acts as a transcriptional factor in mediating the expression of various genes, such as MnSOD, CAT, BIM, BBC3, and CDK6. FOXO3a plays a critical role in the metabolism of bone and cartilage. In this article, we mainly discussed the biological functions of FOXO3a in bone and cartilage diseases, such as osteoporosis (OP), osteoarthritis (OA), rheumatoid arthritis (RA), ankylosing spondylitis (AS), and intervertebral disc degeneration (IDD). FOXO3a can promote osteogenic differentiation, induce osteoblast proliferation, inhibit osteoclast activity, suppress chondrocyte apoptosis, and reduce inflammatory responses. Collectively, up-regulation of FOXO3a expression shows beneficial effects, and FOXO3a has become a potential target for bone and cartilage diseases.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Medical Imaging, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, People’s Republic of China
- First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, People’s Republic of China
| | - Wang Zhan
- First Clinical Medical College, Gannan Medical University, Ganzhou, 341000, People’s Republic of China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, 341000, People’s Republic of China
| | - Luhu Yu
- Department of Clinical Laboratory, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Fang Yu
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Shengrong Bi
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Shiwei Liu
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| | - Guoqiang Yin
- Department of Joint Surgery, Ganzhou Hospital Affiliated to Nanchang University, Ganzhou, 341000, People’s Republic of China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People’s Hospital, Ganzhou, 341000, People’s Republic of China
| |
Collapse
|
18
|
Zhu X, Zhou X, Li S, Liu Z, Yu S, Shi H, Zhu L, Song B, Si Z, Sun M, Zhu W. PFKFB3 decreases α-ketoglutarate production while partial PFKFB3 knockdown in macrophages ameliorates arthritis in tumor necrosis factor-transgenic mice. Int Immunopharmacol 2025; 148:114102. [PMID: 39870011 DOI: 10.1016/j.intimp.2025.114102] [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: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
OBJECTIVE Aberrant 6-phosphofructo-2kinase/fructose-2,6-bisphoshatase 3 (PFKFB3) expression is tightly correlated with multiple steps of tumorigenesis; however, the pathological significance of PFKFB3 in macrophages in patients with rheumatoid arthritis (RA) remains obscure. In this study, we examined whether PFKFB3 modulates macrophage activation and promotes RA development. METHOD Peripheral blood mononuclear cells (PBMCs) from patients with RA, THP-1 cells, and bone marrow-derived macrophages from conditional PFKFB3-knockout mice were used to investigate the mechanism underlying PFKFB3-induced macrophage regulation of RA. RESULT We demonstrated that patients with RA have higher PFKFB3 levels than healthy volunteers. PFKFB3 silencing suppressed M1 macrophage polarization and downregulated IL-1β, CD80, IFIT1, CCL8, and CXCL10 in macrophages of patients with RA. PFKFB3 overexpression markedly upregulated IRF5, HIF1α, IL-1β, CD80, IFI27, IFI44, IFIT1, IFIT3, CCL2, CCL8, CXCL10, CXCL11, and MMP13 in phorbol 12-myristate 13-acetate-induced THP-1 cells, although these changes were partially reversed by PFK15, an inhibitor of PFKFB3 enzyme activity. Co-immunoprecipitation assays revealed that PFKFB3 interacted with GLUD1 and decreased glutamate dehydrogenase (GDH) activity and α-ketoglutarate production. PFKFB3, TNFα, IL-6, IFNγ, CXCL9, CXCL10, CXCL11, MMP13, and MMP19 were downregulated in bone marrow-derived macrophages of conditional PFKFB3-knockout mice relative to those of wild-type mice. Partial PFKFB3 knockdown in macrophages ameliorated the clinical signs of arthritis and bone destruction, inhibited proinflammatory factor expression, and promoted GDH activity and α-ketoglutarate production in tumor necrosis factor-transgenic mice. Single-cell sequencing revealed that macrophages were the most abundant cells in the ankles of arthritic mice, and partial PFKFB3 knockdown promoted M2-like polarization and was correlated with TREM2, SPP1, APOE, and C1Q expression. CONCLUSION PFKFB3 is upregulated in macrophages in patients with RA. PFKFB3 aggravates arthritis by modulating macrophage activity, which may be related to decreased α-ketoglutarate production.
Collapse
Affiliation(s)
- Xiaodong Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Xiaohui Zhou
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shuaiyi Li
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zenghui Liu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Shidi Yu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Hong Shi
- Department of Rheumatology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - LingLing Zhu
- Department of Rheumatology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Baohui Song
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Zihou Si
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Mingshuang Sun
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China
| | - Wei Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, Heilongjiang 157011, China.
| |
Collapse
|
19
|
Miyamoto AT, Shimagami H, Kumanogoh A, Nishide M. Spatial transcriptomics in autoimmune rheumatic disease: potential clinical applications and perspectives. Inflamm Regen 2025; 45:6. [PMID: 39980019 PMCID: PMC11841260 DOI: 10.1186/s41232-025-00369-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Spatial transcriptomics is a cutting-edge technology that analyzes gene expression at the cellular level within tissues while integrating spatial location information. This concept, which combines high-plex RNA sequencing with spatial data, emerged in the early 2010s. Spatial transcriptomics has rapidly expanded with the development of technologies such as in situ hybridization, in situ sequencing, in situ spatial barcoding, and microdissection-based methods. Each technique offers advanced mapping resolution and precise spatial assessments at the single-cell level. Over the past decade, the use of spatial transcriptomics on clinical samples has enabled researchers to identify gene expressions in specific diseased foci, significantly enhancing our understanding of cellular interactions and disease processes. In the field of rheumatology, the complex and elusive pathophysiology of diseases such as rheumatoid arthritis, systemic lupus erythematosus, and Sjögren's syndrome remains a challenge for personalized treatment. Spatial transcriptomics provides insights into how different cell populations interact within disease foci, such as the synovial tissue, kidneys, and salivary glands. This review summarizes the development of spatial transcriptomics and current insights into the pathophysiology of autoimmune rheumatic diseases, focusing on immune cell distribution and cellular interactions within tissues. We also explore the potential of spatial transcriptomics from a clinical perspective and discuss the possibilities for translating this technology to the bedside.
Collapse
Affiliation(s)
- Atsuko Tsujii Miyamoto
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (Ifrec), Osaka University, Suita, Osaka, Japan
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Hiroshi Shimagami
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (Ifrec), Osaka University, Suita, Osaka, Japan
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (Ifrec), Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
- Center for Infectious Diseases for Education and Research (Cider), Osaka University, Suita, Osaka, Japan
- Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS (Camad), Osaka University, Suita, Osaka, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
- Department of Immunopathology, World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (Ifrec), Osaka University, Suita, Osaka, Japan.
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
20
|
Hou FF, Mi JH, Wang Q, Tao YL, Guo SB, Ran GH, Wang JC. Macrophage polarization in sepsis: Emerging role and clinical application prospect. Int Immunopharmacol 2025; 144:113715. [PMID: 39626538 DOI: 10.1016/j.intimp.2024.113715] [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: 10/21/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/15/2024]
Abstract
Sepsis is a severe, potentially fatal condition defined by organ dysfunction due to excessive inflammation. Its complex pathogenesis and poor therapeutic outcomes pose significant challenges in treatment. Macrophages, with their high heterogeneity and plasticity, play crucial roles in both the innate and adaptive immune systems. They can polarize into M1-like macrophages, which promote pro-inflammatory responses, or M2-like macrophages, which mediate anti-inflammatory responses, positioning them as critical mediators in the immune response during sepsis.Macrophages are the main regulators of inflammatory responses, and their polarization is also regulated by inflammatory signaling pathways. This review highlights recent advances in the inflammatory signaling pathways involved in sepsis, mechanism of macrophage polarization mediated by inflammation-related signaling pathways in sepsis, and the role of signaling pathway mediated macrophage polarization in organ dysfunction involved in sepsis. We also explore the therapeutic potential of targeting macrophage polarization for immunotherapy, offering new perspectives on macrophage-targeted treatments for sepsis.
Collapse
Affiliation(s)
- Fei Fei Hou
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China
| | - Jun Hao Mi
- Liuzhou Maternity and Child Healthcare Hospital, Liuzhou 545001, China
| | - Qiong Wang
- Burn and Plastic Surgery Department of Hohhot First Hospital, Hohhot 010030, China
| | - Yan Lin Tao
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China
| | - Shuai Bin Guo
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China
| | - Guang He Ran
- Chongqing Changshou Traditional Cinese Medicine Hospital, 401200 Chongqing, China.
| | - Jing Chao Wang
- Intensive Care Unit Inner Mongolia Medical University Affiliated Hospital, Hohhot 010050, China.
| |
Collapse
|
21
|
Fan Y, Wu YJ, Guo K, Zhou XQ, Abulaiti A, Olatunji OJ, Ji CL, Zuo J. Interaction with IGF1 overrides ANXA2-mediated anti-inflammatory functions of IGFBP5 in vivo. Front Immunol 2025; 15:1539317. [PMID: 39867883 PMCID: PMC11757107 DOI: 10.3389/fimmu.2024.1539317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 01/28/2025] Open
Abstract
Background IGFBP5 is a differentially expressed gene (DEG) between M1 and M2 macrophages. This study explained why it causes opposite effects in different circumstances. Methods Gene expression profiles of various cell subsets were compared by mining a public database. THP-1 cells were treated by siRNAs, recombinant IGFBP5, lipopolysaccharide (LPS), picropodophyllin, IGF1 or the combinations. Clinical implication of IGFBP5 changes was investigated using rheumatoid arthritis (RA) and acute lung injury (ALI) models. IGFBP5-bound and differential proteins were identified by Liquid Chromatography Mass Spectrometry method. Results IGFBP5 situated in the center of a network constructed by the DEGs of M0 and M1/2 macrophages. Its expression negatively correlated to inflammation in vitro. When IGFBP5 was silenced, monocytes released more IL-1β and IL-6. NF-κB downstream proteins were overexpressed. IGFBP5 interacted with ANXA2 directly. In ANXA2-silenced cells, it showed no anti-inflammatory effect. Monocytes of adjuvant-induced arthritis rats and RA patients expressed less IGFBP5 than normal controls, but its blood levels increased significantly. Adipocytes secreted large amounts of IGFBP5. This secretion was reinforced by the above sera. IGFBP5 decreased in ALI mice's blood, while its supplement exacerbated inflammation. By binding to IGF1, IGFBP5 prevented its interaction with IGF1R. An IGF1R inhibitor picropodophyllin antagonized functions of IGF1/IGF1R too, but didn't reinforce the effects of IGFBP5. Conclusion IGFBP5 eases inflammation by interacting with ANXA2, an activator of NF-κB; as an antagonist of IGF1/IGF1R, IGFBP5 may disrupt immune homeostasis in vivo, due to impairment of the latter's anti-inflammatory functions; excessive IGFBP from adipocytes would be a pathogenic factor in certain diseases.
Collapse
Affiliation(s)
- Yan Fan
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Yi-Jin Wu
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
- Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Kai Guo
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Xia-Qing Zhou
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
| | - Abulizi Abulaiti
- Institute of Traditional Chinese Medicine and Ethnic Medicine, Uyghur Medicine Hospital of Akesu Prefecture, Akesu, Xinjiang, China
| | | | - Cong-Lan Ji
- School of Pharmacy, Anhui College of Traditional Chinese Medicine, Wuhu, China
| | - Jian Zuo
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, China
- Department of Pharmacy, The Second Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
22
|
Kim HM, Yoo DH, Kang JW, Lee IC, Bae JS. Anti-Inflammatory Effect of Extract from Fragaria ananassa Duch. Calyx via MAPK and NF-κB Signaling Pathway. J Microbiol Biotechnol 2024; 34:2662-2674. [PMID: 39604003 PMCID: PMC11733547 DOI: 10.4014/jmb.2409.09044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/13/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
Currently, Fragaria ananassa Duch. are discarded as by-products except for the fruit part, so we developed a natural material using the top (= calyx), one of the by-products, and prepared an extract using 70% ethanol to investigate its effects on anti-inflammatory mechanisms. The polyphenol content of 70% ethanol extracts from Fragaria ananassa Duch. calyx was measured to be 265.86 ± 0.85 mg TAE/100 g, respectively. The antioxidant activity was confirmed through the electron donating ability and 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonate) (ABTS) radical scavenging ability measurements. When extracts from Fragaria ananassa Duch. calyx was treated to LPS-induced RAW 264.7 cells, it was confirmed that the production of inflammation-related factors, NO, PGE2, iNOS, COX-2, TNF-a, and IL-6, was inhibited. In addition, it was confirmed that extracts from Fragaria ananassa Duch. calyx affected the MAPK signaling pathway by reducing the protein expression of p-ERK, p-JNK, and p-p38, which are the upper signaling pathways. In addition, it was confirmed to reduce the protein expression of p-p65 and p-IκB, which are NF-κB signaling pathways. Therefore, this study suggests that extracts from Fragaria ananassa Duch. calyx affect the regulation of the production of major inflammation-related factors by inhibiting the MAPK and NF-κB signaling pathway. These results confirmed that extracts from Fragaria ananassa Duch. calyx have the potential to be developed as a new natural material with anti-inflammatory activity.
Collapse
Affiliation(s)
- Hyo-Min Kim
- College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Dan-Hee Yoo
- College of Fusion and Convergence, Seowon University, Cheongju 28674, Republic of Korea
| | - Jung-Wook Kang
- College of Fusion and Convergence, Seowon University, Cheongju 28674, Republic of Korea
| | - In-Chul Lee
- Department of Bio-Cosmetic Science, Seowon University, Cheongju 28674, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea
| |
Collapse
|
23
|
Fang F, Hua M, Yu G. Study of the mechanism of fibroblast-like synoviocytes-derived exosomes inducing macrophages M1 polarization and CD8 +T cells immune regulation ferroptosis and autophagy in rheumatoid arthritis. Immunol Lett 2024; 270:106936. [PMID: 39447763 DOI: 10.1016/j.imlet.2024.106936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that affects the joints. The pathogenesis of RA is complex, involving membrane lipid antioxidant systems, oxidative stress, and lipid peroxidation. In this study, it was found that cysteine dioxygenase 1 (CDO1) is significantly upregulated in RA fibroblast-like synoviocytes (RA-FLS) and that exosomes derived from these RA-FLS deliver CDO1 to promote M1 polarization of macrophages, thus facilitating RA progression. In the immune microenvironment, CD8+T cells play a role in immune regulation by producing cytokines such as interferon gamma (IFNγ) in various diseases. The results of this study suggested that in RA-FLS, CD8+T cells deliver IFNγ, which not only inhibits the viability of RA-FLS but also affects glutathione (GSH) through CDO1, regulating the GPX4 antioxidant signaling pathway to promote ferroptosis and autophagy in cells. It was also discovered that IFNγ enhances the expression of TRI69, ubiquitinates and degrades FSP1, thereby forming a cooperative regulation process of GPX4 and FSP1 in ferroptosis. These findings provide a new direction for the treatment of RA.
Collapse
Affiliation(s)
- Fang Fang
- Department of Immunology, School of Laboratory Medicine, Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, Anhui 233030, China.
| | - Mengqing Hua
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, China
| | - GenMing Yu
- Bengbu Medical College, Bengbu, Anhui 233030, China
| |
Collapse
|
24
|
Cao J, Yang Z. FOXA2 inhibits the TLR4/NF-κB signaling pathway and alleviates inflammatory activation of macrophages in rheumatoid arthritis by repressing LY96 transcription. Cytokine 2024; 184:156796. [PMID: 39486110 DOI: 10.1016/j.cyto.2024.156796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) remains a devastating autoimmune disease characterized by joint damage, inflammation, and disability. This study investigates the function of lymphocyte antigen 96 (LY96) in the inflammatory response in RA and explores its regulatory mechanism. METHODS A mouse model of RA was developed using type II collagen, and the LY96 expression in the ankle joint tissue was determined. Upstream regulators targeting LY96 were investigated using bioinformatics, followed by chromatin immunoprecipitation and luciferase reporter assays for validation. Gain- or loss-of-functions of LY96 and forkhead box A2 (FOXA2) were performed to analyze their roles in arthritis score, pathological changes, and inflammatory responses in mice. The effects of FOXA2 and LY96 on pro-inflammatory activation of macrophages were additionally investigated in vitro using a mouse RAW264.7 macrophage model with lipopolysaccharide treatment. RESULTS LY96 mRNA and protein (MD-2) levels were increased in the RA mice. Knockdown of LY96 alleviated arthritis severity, joint deformities, inflammation, and cartilage destruction in mice. In vitro, the LY96 knockdown reduced the pro-inflammatory activation of RAW264.7 macrophages by inhibiting the TLR4/NF-κB inflammatory signaling transduction. FOXA2 was identified as a transcriptional repressor of LP96 poorly expressed in RA. Overexpression of FOXA2 similarly alleviated inflammation and reduced M1-type macrophages in vivo and in vitro. However, these changes were reversed by the additional LY96 upregulation. CONCLUSION This study suggests that FOXA2 represses LY96 transcription to inhibit the TLR4/NF-κB signaling transduction, thus reducing pro-inflammatory activation of macrophages in the context of RA.
Collapse
Affiliation(s)
- Jin Cao
- Department of Rheumatology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, PR China
| | - Zhaowen Yang
- Department of Rheumatology, Central Hospital Affiliated to Shandong First Medical University, Jinan 250013, Shandong, PR China.
| |
Collapse
|
25
|
Khamis AA, Sharshar AH, Mohamed TM, Abdelrasoul EA, Salem MM. Visnagin alleviates rheumatoid arthritis via its potential inhibitory impact on malate dehydrogenase enzyme: in silico, in vitro, and in vivo studies. GENES & NUTRITION 2024; 19:20. [PMID: 39390383 PMCID: PMC11465529 DOI: 10.1186/s12263-024-00756-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder. The present study aimed to evaluate the in silico, in vitro, and in vivo inhibitory effect of visnagin on malate dehydrogenase activity and elucidate its inflammatory efficacy when combined with methotrexate in the RA rat model. The molecular docking, ADMET simulations, MDH activity, expression, and X-ray imaging were detected. Moreover, CRP, RF, (anti-CCP) antibody, (TNF-α), (IL-6), (IL-17), and (IL-10) were evaluated. The expression levels of MMP3 and FOXP3 genes and CD4, CD25, and CD127 protein levels were assessed. Histological assessment of ankle joints was evaluated. The results revealed that visnagin showed reversible competitive inhibition on MDH with inhibitory constant (Ki) equal to 141 mM with theoretical IC50 equal to 1202.7 mM, LD50 equal to 155.39 mg/kg, and LD25 equal to 77.69 mg/kg. In vivo studies indicated that visnagin exhibited anti-inflammatory effects through decreasing MDH1 activity and expression and induced proliferation of anti-inflammatory CD4+CD25+FOXP3 regulatory T cells with increasing the anti-inflammatory cytokine IL-10 levels. Moreover, visnagin reduced the levels of inflammatory cytokines and the immuno-markers. Our findings elucidate that visnagin exhibits an anti-inflammatory impact against RA through its ability to inhibit the MDH1 enzyme, improve methotrexate efficacy, and reduce oxidative stress.
Collapse
Affiliation(s)
- Abeer A Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| | - Amira H Sharshar
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Tarek M Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Elsayed A Abdelrasoul
- Head Researcher of Special Food and Nutrition Department, Food Technology Research Institute, Agricultural Research Center, Giza, Egypt
| | - Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
26
|
Wang DD, Song MK, Yin Q, Chen WG, Olatunji OJ, Yang K, Zuo J. Qing-Luo-Yin Eased Adjuvant-Induced Arthritis by Inhibiting SIRT1-Controlled Visfatin Production in White Adipose Tissues. J Inflamm Res 2024; 17:6691-6706. [PMID: 39345898 PMCID: PMC11438449 DOI: 10.2147/jir.s474329] [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: 06/19/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Background Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 regulates both metabolism and immune functions. This study investigated if SIRT1 inhibitory property of herbal formula Qing-Luo-Yin (QLY) contributed to its anti-rheumatic effects. Methods Adjuvant-induced arthritis (AIA) rats were treated by QLY and nicotinamide mononucleotide (NMN, a biosynthesis precursor of NAD) for 38 days. After sacrifice, blood, paws, liver and white adipose tissues (WAT) were collected. Pre-adipocytes were cultured by the rats' serum. The medium was used for monocytes culture. Some pre-adipocytes were treated by QLY-derived SIRT1 inhibitors. SIRT1 was silenced or overexpressed beforehand. The samples were subjected to kits-based quantification, polymerase-chain reaction, western-blot, immunofluorescence, and histology experiments. Results AIA rats experienced significant fat loss in liver and WAT. Expression of many SIRT1-related signals like PPARγ, PGC-1α, HSL, ATGL and CPT-1A were altered. QLY attenuated all these abnormalities and joint injuries. By pan-acetylation up-regulation, visfatin was obviously reduced in QLY-treated AIA rats' blood (from 191.8 to 127.0 pg/mL). NMN sustained SIRT1 activation by replenishing NAD, and weakened these effects. QLY-containing serum and the related compounds showed similar impacts on pre-adipocytes, resembling the changes in QLY-treated AIA rats' WAT. These treatments suppressed AIA serum-induced visfatin secretion (from 49.3 to 36.1 and 30.7 pg/mL). This effect was impaired by SIRT1 overexpression. The medium from the compounds-treated pre-adipocytes impaired NF-κB activation in AIA serum-cultured monocytes. Conclusion Besides fat depletion, SIRT1 up-regulation in rheumatic subjects' WAT promotes visfatin production, and exacerbates inflammation. SIRT1 inhibition in WAT is an anti-rheumatic way of QLY independent of immune regulation.
Collapse
Affiliation(s)
- Dan-Dan Wang
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People’s Republic of China
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Meng-Ke Song
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People’s Republic of China
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Qin Yin
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People’s Republic of China
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Wen-Gang Chen
- Department of Pharmacy, the Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | | | - Kui Yang
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Jian Zuo
- Xin’an Medicine Research Center, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, People’s Republic of China
- Research Center of Integration of Traditional Chinese and Western Medicine, Wannan Medical College, Wuhu, 241000, People’s Republic of China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| |
Collapse
|
27
|
Laha A, Nasra S, Bhatia D, Kumar A. Advancements in rheumatoid arthritis therapy: a journey from conventional therapy to precision medicine via nanoparticles targeting immune cells. NANOSCALE 2024; 16:14975-14993. [PMID: 39056352 DOI: 10.1039/d4nr02182g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease that mainly affects the inner lining of the synovial joints and leads to chronic inflammation. While RA is not known as lethal, recent research indicates that it may be a silent killer because of its strong association with an increased risk of chronic lung and heart diseases. Patients develop these systemic consequences due to the regular uptake of heavy drugs such as disease-modifying antirheumatic medications (DMARDs), glucocorticoids (GCs), nonsteroidal anti-inflammatory medicines (NSAIDs), etc. Nevertheless, a number of these medications have off-target effects, which might cause adverse toxicity, and have started to become resistant in patients as well. Therefore, alternative and promising therapeutic techniques must be explored and adopted, such as post-translational modification inhibitors (like protein arginine deiminase inhibitors), RNA interference by siRNA, epigenetic drugs, peptide therapy, etc., specifically in macrophages, neutrophils, Treg cells and dendritic cells (DCs). As the target cells are specific, ensuring targeted delivery is also equally important, which can be achieved with the advent of nanotechnology. Furthermore, these nanocarriers have fewer off-site side effects, enable drug combinations, and allow for lower drug dosages. Among the nanoparticles that can be used for targeting, there are both inorganic and organic nanomaterials such as solid-lipid nanoparticles, liposomes, hydrogels, dendrimers, and biomimetics that have been discussed. This review highlights contemporary therapy options targeting macrophages, neutrophils, Treg cells, and DCs and explores the application of diverse nanotechnological techniques to enhance precision RA therapies.
Collapse
Affiliation(s)
- Anwesha Laha
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Simran Nasra
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| | - Dhiraj Bhatia
- Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar - 382055, Gujarat, India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts & Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad 380009, Gujarat, India.
| |
Collapse
|
28
|
Karpuz M, Aydin HH, Ozgenc E, Erel-Akbaba G, Atlihan-Gundogdu E, Senyigit Z. 99mTc-labeled, tofacitinib citrate encapsulated chitosan microspheres loaded in situ gel formulations for intra-articular treatment of rheumatoid arthritis. Drug Dev Res 2024; 85:e22247. [PMID: 39138857 DOI: 10.1002/ddr.22247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/07/2024] [Accepted: 07/28/2024] [Indexed: 08/15/2024]
Abstract
Inflammatory diseases including rheumatoid arthritis are major health problems. Although different techniques and drugs are clinically available for the diagnosis and therapy of the disease, novel approaches regarding radiolabeled drug delivery systems are researched. Hence, in the present study, it was aimed to design, prepare, and characterize 99mTc-radiolabeled and tofacitinib citrate-encapsulated microsphere loaded poloxamer in situ gel formulations for the intra-articular treatment. Among nine different microsphere formulations, MS/TOFA-9 was chosen as the most proper one due to particle size, high encapsulation efficiency, and in vitro drug release behavior. Poloxamer 338 at a concentration of 15% was used to prepare in situ gel formulations. For intra-articular administration, microspheres were dispersed in an in situ gel containing 15% Poloxamer 338 and characterized in terms of gelation temperature, viscosity, rheological, mechanical, and spreadability properties. After the determination of the safe dose for MS/TOFA-9 and PLX-MS/TOFA-9 as 40 µL/mL in the cell culture study performed on healthy cells, the high anti-inflammatory effects were due to significant cellular inhibition of fibroblasts. In the radiolabeling studies with 99mTc, the optimum radiolabeling condition was determined as 200 ppm SnCl2 and 0.5 mg ascorbic acid, and both 99mTc-MS/TOFA-9 and 99mTc-PLX-MS/TOFA-9 exhibited high cellular binding capacity. In conclusion, although further in vivo experiments are required, PLX-MS/TOFA-9 was found to be a promising agent for intra-articular injection in rheumatoid arthritis.
Collapse
Affiliation(s)
- Merve Karpuz
- Department of Radiopharmacy, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Husniye Hande Aydin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Emre Ozgenc
- Department of Radiopharmacy, Faculty of Pharmacy, Ege University, Izmir, Turkey
| | - Gulsah Erel-Akbaba
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | | | - Zeynep Senyigit
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| |
Collapse
|
29
|
Kiełbowski K, Plewa P, Bratborska AW, Bakinowska E, Pawlik A. JAK Inhibitors in Rheumatoid Arthritis: Immunomodulatory Properties and Clinical Efficacy. Int J Mol Sci 2024; 25:8327. [PMID: 39125897 PMCID: PMC11311960 DOI: 10.3390/ijms25158327] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is a highly prevalent autoimmune disorder. The pathogenesis of the disease is complex and involves various cellular populations, including fibroblast-like synoviocytes, macrophages, and T cells, among others. Identification of signalling pathways and molecules that actively contribute to the development of the disease is crucial to understanding the mechanisms involved in the chronic inflammatory environment present in affected joints. Recent studies have demonstrated that the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway regulates the behaviour of immune cells and contributes to the progression of RA. Several JAK inhibitors, such as tofacitinib, baricitinib, upadacitinib, and filgocitinib, have been developed, and their efficacy and safety in patients with RA have been comprehensively investigated in a number of clinical trials. Consequently, JAK inhibitors have been approved and registered as a treatment for patients with RA. In this review, we discuss the involvement of JAK/STAT signalling in the pathogenesis of RA and summarise the potential beneficial effects of JAK inhibitors in cells implicated in the pathogenesis of the disease. Moreover, we present the most important phase 3 clinical trials that evaluated the use of these agents in patients.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| | - Paulina Plewa
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
| | | | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.)
| |
Collapse
|