1
|
Pelissier A, Laragione T, Harris C, Rodríguez Martínez M, Gulko PS. BACH1 as a key driver in rheumatoid arthritis fibroblast-like synoviocytes identified through gene network analysis. Life Sci Alliance 2025; 8:e202402808. [PMID: 39467637 PMCID: PMC11519322 DOI: 10.26508/lsa.202402808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
RNA-sequencing and differential gene expression studies have significantly advanced our understanding of pathogenic pathways underlying rheumatoid arthritis (RA). Yet, little is known about cell-specific regulatory networks and their contributions to disease. In this study, we focused on fibroblast-like synoviocytes (FLS), a cell type central to disease pathogenesis and joint damage in RA. We used a strategy that computed sample-specific gene regulatory networks to compare network properties between RA and osteoarthritis FLS. We identified 28 transcription factors (TFs) as key regulators central to the signatures of RA FLS. Six of these TFs are new and have not been previously implicated in RA through ex vivo or in vivo studies, and included BACH1, HLX, and TGIF1. Several of these TFs were found to be co-regulated, and BACH1 emerged as the most significant TF and regulator. The main BACH1 targets included those implicated in fatty acid metabolism and ferroptosis. The discovery of BACH1 was validated in experiments with RA FLS. Knockdown of BACH1 in RA FLS significantly affected the gene expression signatures, reduced cell adhesion and mobility, interfered with the formation of thick actin fibers, and prevented the polarized formation of lamellipodia, all required for the RA destructive behavior of FLS. This study establishes BACH1 as a central regulator of RA FLS phenotypes and suggests its potential as a therapeutic target to selectively modulate RA FLS.
Collapse
Affiliation(s)
- Aurelien Pelissier
- IBM Research Europe, Eschlikon, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Teresina Laragione
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolyn Harris
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Percio S Gulko
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
2
|
Zhang J, Xie X, Shen Q, Yuan C, Lu G, Xiao W, Gong W, Fu X, Feng X. Rhaponticin Alleviates Collagen-induced Arthritis by Inhibiting NLRP3/GSDMD-mediated Neutrophil Extracellular Traps. Inflammation 2024:10.1007/s10753-024-02228-7. [PMID: 39725843 DOI: 10.1007/s10753-024-02228-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Neutrophil extracellular traps (NETs) play an important role in the inflammatory response and progressive joint destruction in rheumatoid arthritis (RA). Rhaponticin (Rha) is a stilbene glycoside compound with antioxidant and anti-inflammatory effects. This study aimed to investigate the therapeutic potential of Rha in RA, with a specific focus on its effects on NETs and on the underlying mechanisms of Rha. NETs formation induced by phorbol 12-myristate 13-acetate (PMA) and a collagen-induced arthritis (CIA) mouse model were implemented to evaluate the pharmacological effects of Rha in vitro and in vivo. The potential mechanism of Rha in improving RA was screened and verified using the SuperPred and DisGeNET databases. Disulfiram (a GSDMD inhibitor) and S100a8cre GSDMDfl/fl mice were used to confirm whether GSDMD is key to the role of Rha. The findings demonstrate that Rha significantly inhibited reactive oxygen species and NETs production in PMA-activated neutrophils. In vivo, Rha treatment significantly relieved joint symptoms in CIA mice and NETs production. Mechanistically, Rha reduced NETs production via inhibition of NLRP3/GSDMD activation. Neutrophil-specific GSDMD depletion eliminated the effects of Rha on NETs production in vitro. Disulfiram eliminated the effects of Rha on the inhibition of NETs production and alleviated joint inflammation in mice in vivo and in vitro. Overall, our results indicated that Rha exerts a protective effect against CIA by inhibiting NETs production through the NLRP3/GSDMD pathway. The results of this study provide new strategies for treating RA.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Xinyue Xie
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China
| | - Qinhao Shen
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Chenchen Yuan
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Guotao Lu
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Weiming Xiao
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Weijuan Gong
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China
| | - Xiaoyan Fu
- Department of Rehabilitation, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Yangzhou University, Kunshan, 215300, China.
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
| |
Collapse
|
3
|
Mathew S, Ashraf S, Shorter S, Tozzi G, Koutsikou S, Ovsepian SV. Neurobiological Correlates of Rheumatoid Arthritis and Osteoarthritis: Remodelling and Plasticity of Nociceptive and Autonomic Innervations in Synovial Joints. Neuroscientist 2024:10738584241293049. [PMID: 39668598 DOI: 10.1177/10738584241293049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Swelling, stiffness, and pain in synovial joints are primary hallmarks of osteoarthritis and rheumatoid arthritis. Hyperactivity of nociceptors and excessive release of inflammatory factors and pain mediators play a crucial role, with emerging data suggesting extensive remodelling and plasticity of joint innervations. Herein, we review structural, functional, and molecular alterations in sensory and autonomic axons wiring arthritic joints and revisit mechanisms implicated in the sensitization of nociceptors, leading to chronic pain. Sprouting and reorganization of sensory and autonomic fibers with the invasion of ectopic branches into surrounding inflamed tissues are associated with the upregulation of pain markers. These changes are frequently complemented by a phenotypic switch of sensory and autonomic profiles and activation of silent axons, inferring homeostatic adjustments and reprogramming of innervations. Identifying critical molecular players and neurobiological mechanisms underpinning the rewiring and sensitization of joints is likely to elucidate causatives of neuroinflammation and chronic pain, assisting in finding new therapeutic targets and opportunities for interventions.
Collapse
Affiliation(s)
- Sharon Mathew
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
| | - Sadaf Ashraf
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Susan Shorter
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
| | - Gianluca Tozzi
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
| | - Stella Koutsikou
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
- Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham Maritime, Kent, UK
| | - Saak V Ovsepian
- Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, Kent, UK
- Faculty of Medicine, Tbilisi State University, Tbilisi, Republic of Georgia
| |
Collapse
|
4
|
Guo Q, Li J, Wang J, Li L, Wei J, Zhang L. The advent of chimeric antigen receptor T Cell therapy in recalibrating immune balance for rheumatic autoimmune disease treatment. Front Pharmacol 2024; 15:1502298. [PMID: 39734406 PMCID: PMC11672202 DOI: 10.3389/fphar.2024.1502298] [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: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
CAR-T cell therapy, a cutting-edge cellular immunotherapy with demonstrated efficacy in treating hematologic malignancies, also exhibits significant promise for addressing autoimmune diseases. This innovative therapeutic approach holds promise for achieving long-term remission in autoimmune diseases, potentially offering significant benefits to affected patients. Current targets under investigation for the treatment of these conditions include CD19, CD20, and BCMA, among others. However, CAR-T therapy faces difficulties such as time-consuming cell manufacturing, complex and expensive process, and the possibility of severe adverse reactions complicating the treatment, etc. This article examines CAR-T therapy across various rheumatic autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS), systemic sclerosis (SSc), antisynthetase syndrome (ASS), and ANCA-associated vasculitis (AAV), highlighting both therapeutic advancements and ongoing challenges.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Jie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Juanjuan Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Linxin Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jia Wei
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
5
|
Li N, Yi YK, Zhao J, Wang Q, Yu JY, You YT, Zhu YY, Liu YY, Zhao XS, Pan DM. Kaempferol Improved Rheumatoid Arthritis by Regulating the Immune Imbalance of Treg/Th17. Curr Med Sci 2024; 44:1259-1269. [PMID: 39673582 DOI: 10.1007/s11596-024-2925-8] [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: 05/12/2023] [Accepted: 08/18/2024] [Indexed: 12/16/2024]
Abstract
OBJECTIVE The objective of this study was to explore the therapeutic effects of kaempferol (Kae) on rheumatoid arthritis (RA) and to elucidate the underlying mechanisms. METHODS The collagen-induced arthritis (CIA) model was established using collagen II to induce RA. Mice were treated with Kae at a dose of 25 or 50 mg/kg/day via gavage. Pathological changes in the ankle joint were analyzed. Enzyme-linked immunosorbent assay (ELISA) was employed to measure the levels of inflammatory factors. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was used to assess the expression of genes associated with the balance of regulatory T (Treg)/T helper 17 (Th17) cells. Flow cytometry was utilized to determine the Treg/Th17 ratio. Furthermore, these techniques were employed to evaluate the impact of miR-34a and Foxp3 dysregulation on cellular functions in RA under the influence of Kae. Dual luciferase reporter assay was conducted to analyze the binding of miR-34a to Foxp3. RESULTS Treatment with Kae led to a downregulation of receptor-related orphan receptor gamma t (RORγt) and IL-17 expression, and an upregulation of Foxp3, IL-10, and TGF-β expression in CIA mice. Kae intervention inhibited the production of proinflammatory cytokines and increased the production of anti-inflammatory cytokines. Furthermore, Kae treatment suppressed the expression of miR-34a, which was identified as a target of miR-34a. Finally, Kae regulated Treg/ Th17 balance-related genes and cellular inflammation through the miR-34a/Foxp3 axis. CONCLUSION The study demonstrated that Kae effectively ameliorates CIA in mice by modulating the Treg/Th17 balance and related genes via the miR-34a/Foxp3 axis. These findings suggest that Kae may serve as a promising therapeutic agent for the treatment of RA and for restoring immune homeostasis.
Collapse
MESH Headings
- Animals
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/genetics
- Mice
- Kaempferols/pharmacology
- Kaempferols/administration & dosage
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- MicroRNAs/genetics
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Male
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Mice, Inbred DBA
- Disease Models, Animal
- Cytokines/metabolism
- Cytokines/genetics
- Interleukin-10/genetics
- Interleukin-10/metabolism
Collapse
Affiliation(s)
- Nan Li
- Department of Clinical Basis of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yan-Kui Yi
- Department of Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Jie Zhao
- Health College of Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qiang Wang
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Guangzhou, 529500, China
| | - Jie-Ying Yu
- Department of Clinical Basis of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan-Ting You
- Department of Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yong-Yan Zhu
- Department of Chemistry and Analysis of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Yan-Yan Liu
- Department of Internal Medicine of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Xiao-Shan Zhao
- Department of Syndrome Laboratory of Integrated Chinese and Western Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| | - Dong-Mei Pan
- Department of Clinical Basis of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
6
|
Bhuvaneshwari S, Venkataraman K, Sankaranarayanan K. Exploring potential ion channel targets for rheumatoid arthritis: combination of network analysis and gene expression analysis. Biotechnol Appl Biochem 2024; 71:1405-1427. [PMID: 39049164 DOI: 10.1002/bab.2638] [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: 12/28/2023] [Accepted: 06/29/2024] [Indexed: 07/27/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the synovial membrane that leads to the destruction of cartilage and bone. Currently, pharmacological targeting of ion channels is being increasingly recognized as an attractive and feasible strategy for the treatment of RA. The present work employs a network analysis approach to predict the most promising ion channel target for potential RA-treating drugs. A protein-protein interaction map was generated for 343 genes associated with inflammation in RA and ion channel genes using Search Tool for the Retrieval of Interacting Genes and visualized using Cytoscape. Based on the betweenness centrality and traffic values as key topological parameters, 17 hub nodes were identified, including FOS (9800.85), tumor necrosis factor (3654.60), TGFB1 (3305.75), and VEGFA (3052.88). The backbone network constructed with these 17 hub genes was intensely analyzed to identify the most promising ion channel target using network analyzer. Calcium permeating ion channels, especially store-operated calcium entry channels, and their associated regulatory proteins were found to highly interact with RA inflammatory hub genes. This significant ion channel target for RA identified by theoretical and statistical studies was further validated by a pilot case-control gene expression study. Experimental verification of the above findings in 75 RA cases and 25 controls showed increased ORAI1 expression. Thus, with a combination of network analysis approach and gene expression studies, we have explored potential targets for RA treatment.
Collapse
Affiliation(s)
- Sampath Bhuvaneshwari
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| | | | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology, Anna University, Chennai, India
| |
Collapse
|
7
|
Xian Y, Ye J, Tang Y, Zhang N, Peng C, Huang W, He G. Deubiquitinases as novel therapeutic targets for diseases. MedComm (Beijing) 2024; 5:e70036. [PMID: 39678489 PMCID: PMC11645450 DOI: 10.1002/mco2.70036] [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: 07/10/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
Deubiquitinating enzymes (DUBs) regulate substrate ubiquitination by removing ubiquitin or cleaving within ubiquitin chains, thereby maintaining cellular homeostasis. Approximately 100 DUBs in humans counteract E3 ubiquitin ligases, finely balancing ubiquitination and deubiquitination processes to maintain cellular proteostasis and respond to various stimuli and stresses. Given their role in modulating ubiquitination levels of various substrates, DUBs are increasingly linked to human health and disease. Here, we review the DUB family, highlighting their distinctive structural characteristics and chain-type specificities. We show that DUB family members regulate key signaling pathways, such as NF-κB, PI3K/Akt/mTOR, and MAPK, and play crucial roles in tumorigenesis and other diseases (neurodegenerative disorders, cardiovascular diseases, inflammatory disorders, and developmental diseases), making them promising therapeutic targets Our review also discusses the challenges in developing DUB inhibitors and underscores the critical role of the DUBs in cellular signaling and cancer. This comprehensive analysis enhances our understanding of the complex biological functions of the DUBs and underscores their therapeutic potential.
Collapse
Affiliation(s)
- Yali Xian
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Jing Ye
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Yu Tang
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine ResourcesSchool of PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Gu He
- Department of Dermatology & VenerologyState Key Laboratory of BiotherapyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
8
|
Cao Y, Zhou M, Xu T. Causal Effects of Rheumatoid Arthritis, Ankylosing Spondylitis, Juvenile Idiopathic Arthritis on Psoriasis: A Mendelian Randomization Study. Clin Cosmet Investig Dermatol 2024; 17:2583-2593. [PMID: 39583152 PMCID: PMC11585978 DOI: 10.2147/ccid.s490250] [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: 08/22/2024] [Accepted: 11/15/2024] [Indexed: 11/26/2024]
Abstract
Background It is well-documented that rheumatoid arthritis (RA), ankylosing spondylitis (AS), and juvenile idiopathic arthritis (JIA) often exhibit skin manifestations, with psoriasis typically occurring around the time of diagnosis. Thus, it is essential to investigate the potential causal relationship between these forms of arthritis and psoriasis. Methods The OpenGWAS provided traitIDs for exposure factors (RA (bbj-A-74), AS (ebi-A-GCST005529), and JIA (finn-b-JUVEN-ARTHR)) and outcome (psoriasis, finn-b-L12-PSORIASIS). bbj-A-74 had 19,190 samples (9,739,303 SNPs), ebi-A-GCST005529 had 22,647 samples (99,962 SNPs), finn-b-JUVEN-ARTHR had 173,622 samples (16,380,296 SNPs), and psoriasis had 216,752 samples (16,380,464 SNPs). Initially, 57 RA SNPs, 25 AS SNPs, and 5 JIA SNPs were acquired. Causal links were explored via univariate Mendelian Randomization (UVMR) analysis, with sensitivity analyses ensuring reliability. Additionally, multivariate MR (MVMR) analysis was conducted to further estimate the effect of each exposure factor on psoriasis. Results Significant causal links (P < 0.05, OR > 1) were found between bbj-A-74, ebi-A-GCST005529, finn-b-JUVEN-ARTHR, and finn-b-L12-PSORIASIS, indicating associations of RA, AS, and JIA with psoriasis. Sensitivity analyses ensured the reliability of these finding, showing no heterogeneity, horizontal pleiotropy, or SNP locus oversensitivity in UVMR results. Furthermore, MVMR analysis revealed AS and JIA as psoriasis risk factors, while RA showed non-significant protective effects. This suggests AS and JIA may contribute to psoriasis onset or exacerbation when coexisting. Conclusion MR analyses were conducted to investigate the causal links between RA, AS, JIA, and psoriasis, enhancing our grasp of the underlying mechanisms of psoriasis.
Collapse
Affiliation(s)
- Yongping Cao
- Department of Dermatology, Hangzhou Third People’s Hospital, Hangzhou City, Zhejiang Province, People’s Republic of China
| | - Mengyun Zhou
- Department of Dermatology, Hangzhou Third People’s Hospital, Hangzhou City, Zhejiang Province, People’s Republic of China
| | - Tianhong Xu
- Department of Dermatology, Hangzhou Third People’s Hospital, Hangzhou City, Zhejiang Province, People’s Republic of China
| |
Collapse
|
9
|
Liao H, Zheng J, Lu J, Shen HL. NF-κB Signaling Pathway in Rheumatoid Arthritis: Mechanisms and Therapeutic Potential. Mol Neurobiol 2024:10.1007/s12035-024-04634-2. [PMID: 39560902 DOI: 10.1007/s12035-024-04634-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune chronic inflammatory disease that imposes a heavy economic burden on patients and society. Bone and cartilage destruction is considered an important factor leading to RA, and inflammation, oxidative stress, and mitochondrial dysfunction are closely related to bone erosion and cartilage destruction in RA. Currently, there are limitations in the clinical treatment methods for RA, which urgently necessitates finding new effective treatments for patients. Nuclear transcription factor-κB (NF-κB) is a signaling transcription factor that is widely present in various cells. It plays an important role as a stress source in the cellular environment and regulates gene expression in processes such as immunity, inflammation, cell proliferation, and apoptosis. NF-κB has long been recognized as a pathogenic factor of RA, and its activation can exacerbate RA by promoting inflammation, oxidative stress, mitochondrial dysfunction, and bone destruction. Conversely, inhibiting the activity of the NF-κB pathway effectively inhibits these pathological processes, thereby alleviating RA. Therefore, NF-κB may be a potential therapeutic target for RA. This article describes the physiological structure of NF-κB and its important role in RA through the regulation of oxidative stress, inflammatory response, mitochondrial function, and bone destruction. Meanwhile, we also summarized the impact of NF-κB crosstalk with other signaling pathways on RA and the effect of related drugs or inhibitors targeting NF-κB on RA. The purpose of this article is to provide evidence for the role of NF-κB in RA and to emphasize its significant role in RA by elucidating the mechanisms, so as to provide a theoretical basis for targeting the NF-κB pathway as a treatment for RA.
Collapse
Affiliation(s)
- Haiyang Liao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jianxiong Zheng
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Jinyue Lu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China
| | - Hai-Li Shen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, People's Republic of China.
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, 730000, People's Republic of China.
| |
Collapse
|
10
|
Luo Y, Jiang LY, Liao ZZ, Wang YY, Wang YD, Xiao XH. Metabolic Regulation of Inflammation: Exploring the Potential Benefits of Itaconate in Autoimmune Disorders. Immunology 2024. [PMID: 39542834 DOI: 10.1111/imm.13875] [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: 09/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
Itaconic acid and its metabolites have demonstrated significant therapeutic potential in various immune diseases. Originating from the tricarboxylic acid cycle in immune cells, itaconic acid can modulate immune responses, diminish inflammation, and combat oxidative stress. Recent research has uncovered multiple mechanisms through which itaconic acid exerts its effects, including the inhibition of inflammatory cytokine production, activation of anti-inflammatory pathways, and modulation of immune cell function by regulating cellular metabolism. Cellular actions are influenced by the modulation of metabolic pathways, such as inhibiting succinate dehydrogenase (SDH) activity or glycolysis, activation of nuclear-factor-E2-related factor 2 (Nrf2), boosting cellular defences against oxidative stress, and suppression of immune cell inflammation through the NF-κB pathway. This comprehensive review discusses the initiation, progression, and mechanisms of action of itaconic acid and its metabolites, highlighting their modulatory effects on various immune cell types. Additionally, it examines their involvement in immune disease like rheumatoid arthritis, multiple sclerosis, type 1 diabetes mellitus, and autoimmune hepatitis, offering greater understanding for creating new therapies for these ailments.
Collapse
Affiliation(s)
- Yin Luo
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li-Yan Jiang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhe-Zhen Liao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuan-Yuan Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ya-Di Wang
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin-Hua Xiao
- The First Affiliated Hospital, Department of Metabolism and Endocrinology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
11
|
Shi Y, Lei Y, Guo S, Li L, Li X, Liu X, Ding S. Screening Anti-Rheumatoid Arthritis Synovitis Effective Ingredients of Total Flavonoid From Artemisia argyi Folium Based on Spectrum-Effect Relationship. PHYTOCHEMICAL ANALYSIS : PCA 2024. [PMID: 39532485 DOI: 10.1002/pca.3479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Flavonoids are the main nonvolatile component responsible for the anti-rheumatoid arthritis (RA) synovitis activities of Artemisia argyi Folium. However, the effective ingredient remains unidentified. Spectrum-effect relationships analysis was a reliable and efficient strategy for herbal effective ingredient discovery. OBJECTIVES This study aimed to screen the effective ingredients within the total flavonoid from Artemisia argyi Folium (TFAA) that exhibit anti-RA synovitis activities based on spectrum-effect relationship. METHODS TFAA was obtained through ethanol extraction and subsequent purification with D101 resin from 15 distinct batches of Artemisia argyi Folium. The fingerprint of TFAA was established using HPLC, and its efficacy against RA synovitis was evaluated by determining the inhibition rate of nitric oxide (NO) on MH7A synovioblast induced by TNF-α. Common peaks were identified using HPLC-MS/MS and authentic standards. The spectrum-effect relationships between the fingerprints and efficacy were analyzed by gray relational analysis (GRA), canonical correlation analysis (CCA), and partial least squares regression analysis (PLSR) to pinpoint the peaks responsible for the anti-RA synovitis activity, and the results were further verified by in vitro anti-RA synovitis experiments and molecular docking studies. RESULTS The fingerprint revealed 14 common peaks, and 12 compounds were identified, including four caffeoylquinic acids and eight flavonoids. Among them, five flavonoids-X10 (hispidulin), X11 (jaceosidin), X12 (centaureidin), X13 (eupatilin), and X14 (casticin)-were highly relevant to anti-RA synovitis activity. Verification experiments confirmed their inhibitory effect on NO production and cytokine secretion in MH7A cells, showing anti-RA synovitis potential, which was consistent with the established spectrum effect relationship. The underlying mechanism might be related to the inhibition of iNOS. CONCLUSION Hispidulin, jaceosidin, centaureidin, eupatilin, and casticin were the key effective ingredient of TFAA responsible for its anti-RA synovitis effects. These compounds can serve as quality control markers for Artemisia argyi Folium and as lead compounds for anti-RA synovitis treatment.
Collapse
Affiliation(s)
- Yu Shi
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Yueyue Lei
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Shiwen Guo
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Lujun Li
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
- Gansu Herbal Medicine Planting Co., Ltd, Lanzhou, China
| | - Xu Li
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Xinyi Liu
- National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Key Laboratory of Biotechnology of Chinese Traditional Medicine, College of Health Science and Engineering, Hubei University, Wuhan, China
| | - Shuiping Ding
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Lan S, Jin S, Lin R, Chen B, Gao F, Zhang S, Chen L, Li H. Effects of green tea based oral health strategies on disease activity in rheumatoid arthritis. Front Med (Lausanne) 2024; 11:1413753. [PMID: 39564506 PMCID: PMC11573505 DOI: 10.3389/fmed.2024.1413753] [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: 04/07/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024] Open
Abstract
Aim In this study, we aimed to evaluate oral health strategies in rheumatoid arthritis (RA) patients with periodontitis. Methods We enrolled 110 RA patients with periodontitis who were diagnosed in a Grade A tertiary hospital into an oral health strategies program. The control and test groups comprised 55 cases each. The management effect was evaluated by self-care ability, oral health-related quality of life, RA-related clinical indicators, and the DAS28 score. The control group received routine nursing, whereas the test group was in a self-health management program for 3 months. Results After the intervention, compared to the control group, the test group showed better self-care ability, oral health-related quality of life score, RA-related clinical indicators, and DAS28 score (P < 0.05). Conclusion Our oral health strategies program slowed down the progression of the disease and can be popularized in patients.
Collapse
Affiliation(s)
- Sanrong Lan
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- The School of Nursing, Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
| | - Shuang Jin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Rong Lin
- The School of Nursing, Fujian Medical University, Fuzhou, China
| | - Baochuan Chen
- Fujian Medical University 2nd Affiliated Hospital, Quanzhou, China
| | - Fei Gao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
| | - Shengli Zhang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
| | - Lili Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- The School of Nursing, Fujian Medical University, Fuzhou, China
- Fujian Provincial Hospital, Fuzhou, China
| | - Hong Li
- The School of Nursing, Fujian Medical University, Fuzhou, China
| |
Collapse
|
13
|
Kirdaite G, Denkovskij J, Mieliauskaite D, Pachaleva J, Bernotiene E. The Challenges of Local Intra-Articular Therapy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1819. [PMID: 39597004 PMCID: PMC11596802 DOI: 10.3390/medicina60111819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Fibroblast-like synoviocytes (FLSs) are among the main disease-driving players in most cases of monoarthritis (MonoA), oligoarthritis, and polyarthritis. In this review, we look at the characteristics and therapeutic challenges at the onset of arthritis and during follow-up management. In some cases, these forms of arthritis develop into autoimmune polyarthritis, such as rheumatoid arthritis (RA), whereas local eradication of the RA synovium could still be combined with systemic treatment using immunosuppressive agents. Currently, the outcomes of local synovectomies are well studied; however, there is still a lack of a comprehensive analysis of current local intra-articular treatments highlighting their advantages and disadvantages. Therefore, the aim of this study is to review local intra-articular therapy strategies. According to publications from the last decade on clinical studies focused on intra-articular treatment with anti-inflammatory molecules, a range of novel slow-acting forms of steroidal drugs for the local treatment of synovitis have been investigated. As pain is an essential symptom, caused by both inflammation and cartilage damage, various molecules acting on pain receptors are being investigated in clinical trials as potential targets for local intra-articular treatment. We also overview the new targets for local treatment, including surface markers and intracellular proteins, non-coding ribonucleic acids (RNAs), etc.
Collapse
Affiliation(s)
- Gailute Kirdaite
- Department of Personalised Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jaroslav Denkovskij
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
| | - Diana Mieliauskaite
- Department of Personalised Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Jolita Pachaleva
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
| | - Eiva Bernotiene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania (E.B.)
- Faculty of Fundamental Sciences, Vilnius Gediminas Technical University, VilniusTech, Sauletekio al. 11, LT-10223 Vilnius, Lithuania
| |
Collapse
|
14
|
Zhang Y, Yu H, Fu J, Zhuo R, Xu J, Liu L, Dai M, Li Z. Oxidative balance score and the potential for suffering rheumatoid arthritis: a cross-sectional study. Front Immunol 2024; 15:1454594. [PMID: 39555070 PMCID: PMC11563821 DOI: 10.3389/fimmu.2024.1454594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024] Open
Abstract
Objective Our study was conducted to explore the link between oxidative balance score (OBS) and rheumatoid arthritis (RA). Methods A total of 21,415 participants were included in our research from five cycles (2011-2012, 2013-2014, 2015-2016, 2017-2018, and 2017-2020) of the National Health and Nutrition Examination Survey (NHANES). Moreover, 20 elements related to diet as well as lifestyle were combined to calculate OBS. The relationship between OBS and RA was assessed by employing multivariable regression analysis, and further exploration was carried out through subgroup analysis, restricted cubic spline analysis, and sensitivity analysis. Multiple covariates were selected to adjust the model for more robust results. Results In our cross-sectional study, a higher OBS has a protective effect on the development of RA (OR = 0.98, 95% CI: 0.97 to 0.99). In contrast to individuals aged ≥60, the result is more prominent in the population aged 20-60 (OR = 0.97, 95% CI: 0.96 to 0.98). Marital status appears to introduce interference in the relationship between OBS and RA, and unmarried individuals exhibited different outcomes (OR = 1.02, 95% CI: 0.99 to 1.04) compared to others. The positive influence of OBS was more evident in patients with chronic kidney disease and cardiovascular disease, while it was stronger in individuals without diabetes and liver disease. Conclusion A higher OBS correlates with a reduced odd of RA. Further studies are needed to shoot more sights on improving dietary habits and lifestyles to gain proper OBS and explore whether OBS can be one of the measurements utilized to measure the risk of RA.
Collapse
Affiliation(s)
- Yimin Zhang
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| | - Hao Yu
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Urology, The Second Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jianfei Fu
- Department of Medical Records and Statistics, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Renjie Zhuo
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| | - Jin Xu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| | - Liya Liu
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| | - Manyun Dai
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| | - Zhen Li
- School of Public Health, Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
15
|
Liu Y, Qu Y, Liu C, Zhang D, Xu B, Wan Y, Jiang P. Neutrophil extracellular traps: Potential targets for the treatment of rheumatoid arthritis with traditional Chinese medicine and natural products. Phytother Res 2024; 38:5067-5087. [PMID: 39105461 DOI: 10.1002/ptr.8311] [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: 02/27/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. Abnormal formation of neutrophil extracellular traps (NETs) at the synovial membrane leads to the release of many inflammatory cytokines, including IL-1β, IL-6, and TNF-α. Elastase, histone H3, and myeloperoxidase, which are carried by NETs, damage the soft tissues of the joints and aggravate the progression of RA. The balance of NET formation coordinates the pro-inflammatory and anti-inflammatory effects and plays a key role in the development of RA. Therefore, when NETs are used as effector targets, highly targeted drugs with fewer side effects can be developed to treat RA without damaging the host immune system. Currently, an increasing number of studies have shown that traditional Chinese medicines and natural products can regulate the formation of NETs through multiple pathways to counteract RA, which shows great potential for the treatment of RA and has a promising future for clinical application. In this article, we review the latest biological progress in understanding NET formation, the mechanism of NETs in RA, and the potential targets or pathways related to the modulation of NET formation by Chinese medicines and natural products. This review provides a relevant basis for the use of Chinese medicines and natural products as natural adjuvants in the treatment of RA.
Collapse
Affiliation(s)
- Yuan Liu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Yuan Qu
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
| | - Chuanguo Liu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Di Zhang
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bing Xu
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yakun Wan
- School of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ping Jiang
- The first Clinical Medical College, Shandong University of Chinese Traditional Medicine, Jinan, China
- Rheumatology and Immunology Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
16
|
Gaddis JM, Arellano E, Bialaszewski R, Chau-Zanetti D, Torres T, Bergman E, Baker K, Gadad B. Prevalence and Economic Burden of Osteoarthritis and Rheumatoid Arthritis in the Medically Underserved Rio Grande Valley: A Retrospective Longitudinal Analysis. Cureus 2024; 16:e74521. [PMID: 39726504 PMCID: PMC11671120 DOI: 10.7759/cureus.74521] [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] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Introduction Osteoarthritis and rheumatoid arthritis, two of the most common forms of arthritis worldwide, are chronic diseases affecting the joints. The Rio Grande Valley is characterized by an abundance of health disparities, with previous studies showing increased rates of multiple diseases and disorders in this region. This study aimed to determine both the prevalence and the risk-adjusted average cost of osteoarthritis and rheumatoid arthritis in the Rio Grande Valley and to compare them with the national average. We hypothesize that the prevalence and risk-adjusted average cost of osteoarthritis and rheumatoid arthritis in the Rio Grande Valley will be greater than the corresponding national averages. Methods Publicly accessible Medicare beneficiary data were utilized for our retrospective longitudinal, observational study. Osteoarthritis and rheumatoid arthritis data for risk-adjusted average total cost, overall prevalence, sex-stratified prevalence, and ethnicity-stratified prevalence, from 2012 to 2022, were compared between the Rio Grande Valley and the national means using specific domains of the "Mapping Medicare Disparities by Population" tool. Independent t-tests and a Mann-Whitney U test compared prevalence rates and risk-adjusted average total cost means, respectively, between the Rio Grande Valley and the national averages. Results Overall, the prevalence of osteoarthritis and rheumatoid arthritis in the Rio Grande Valley was significantly higher than the national average (39.9% vs. 26.9%, p < 0.001). Women in the region exhibited significantly higher rates of osteoarthritis and rheumatoid arthritis compared to the national average (47.6% vs. 32.1%, p < 0.001), and a similar trend was seen among Hispanic residents (41.6%) compared to the national mean (32.1%) (p < 0.001). Furthermore, the risk-adjusted average total cost for individuals residing in the Rio Grande Valley ($16,084.40) significantly exceeded the risk-adjusted average total cost nationally ($13,073.90) (p < 0.001). Conclusion In the Rio Grande Valley, there is an increased prevalence of osteoarthritis and rheumatoid arthritis compared to the national mean of Medicare beneficiary patients, particularly in women and those of Hispanic heritage. The substantial increase in risk-adjusted average total cost to treat osteoarthritis and rheumatoid arthritis highlights the economic burden faced by residents in the region.
Collapse
Affiliation(s)
- John M Gaddis
- Orthopedic Surgery, The University of Texas Rio Grande Valley School of Medicine, Edinburg, USA
| | - Elias Arellano
- Internal Medicine, The University of Texas Rio Grande Valley School of Medicine, Edinburg, USA
| | - Ryan Bialaszewski
- School of Medicine, The University of Texas Rio Grande Valley, Edinburg, USA
| | - Dominic Chau-Zanetti
- Internal Medicine, The University of Texas Rio Grande Valley School of Medicine, Edinburg, USA
| | - Tyler Torres
- General Surgery, The University of Texas Rio Grande Valley School of Medicine, Edinburg, USA
| | | | - Kelsey Baker
- Neuroscience, The University of Texas Rio Grande Valley, Edinburg, USA
| | - Bharathi Gadad
- Psychiatry and Behavioral Sciences, The University of Texas Rio Grande Valley School of Medicine, Edinburg, USA
| |
Collapse
|
17
|
Qiu S, Zhu F, Tong L. Application of targeted drug delivery by cell membrane-based biomimetic nanoparticles for inflammatory diseases and cancers. Eur J Med Res 2024; 29:523. [PMID: 39472940 PMCID: PMC11523786 DOI: 10.1186/s40001-024-02124-8] [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: 08/02/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Drug-carrying nanoparticles can be recognized and captured by macrophages and cleared away by the immune system, resulting in reduced drug efficacy and representing the main drawbacks. Biomimetic nanoparticles, which are coated with cell membranes from natural resources, have been applied to address this problem. This type of nanoparticle maintains some specific biological activities, allowing them to carry drugs reaching designated tissues effectively and have a longer time in circulation. This review article aims to summarize recent progress on biomimetic nanoparticles based on cell membranes. In this paper, we have introduced the classification of biomimetic nanoparticles, their preparation and characterization, and their applications in inflammatory diseases and malignant tumors. We have also analyzed the shortcomings and prospects of this technology, hoping to provide some clues for basic researchers and clinicians engaged in this field.
Collapse
Affiliation(s)
- Shijie Qiu
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, Heilongjiang Province, China
| | - Feifan Zhu
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, Heilongjiang Province, China
| | - Liquan Tong
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, Heilongjiang Province, China.
| |
Collapse
|
18
|
Tan YK, Thumboo J. Understanding Ultrasound Power Doppler Synovitis at Clinically Quiescent Joints and Thermographic Joint Inflammation Assessment in Patients with Rheumatoid Arthritis. Diagnostics (Basel) 2024; 14:2384. [PMID: 39518352 PMCID: PMC11545627 DOI: 10.3390/diagnostics14212384] [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: 10/05/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory arthritis. We aim to study subclinical PD synovitis and thermographic joint inflammation assessment in patients with RA. Methods: We compared (1) PD synovitis at clinically quiescent (non-swollen; non-tender) joints based on patients' disease activity and (2) thermography (hands/wrists) outcomes based on PD joint inflammation findings and patient's disease activity. Results: Among eighty RA patients (mean (SD) age 57.0 (12.6) years; 61 of whom (76.3%) were female), the wrists (62.7%), second metacarpophalangeal joints (MCPJs) (37.0%), third MCPJs (33.8%), fourth MCPJs (24.8%), and fifth MCPJs (20.9%) were the five joint sites most frequently displaying subclinical PD synovitis; with no statistically significance differences (p > 0.05) between patients with 28-joint disease activity score (DAS28) < 3.2 versus those with DAS28 ≥ 3.2. At these five joint sites bilaterally, (1) the total maximum (Total Tmax), total average (Total Tavg), and total minimum (Total Tmin) temperatures were significantly greater (p < 0.05) for Total PD (TPD) score >1 versus TPD score ≤ 1, while their area under the ROC curve (AUC) values in identifying TPD score >1 ranged from 0.789 to 0.810, and (2) Total Tmax, Total Tavg, Total Tmin, and TPD score were significantly greater (p < 0.05) for patients with DAS28 ≥ 3.2 versus those with DAS28 < 3.2. Conclusions: Our results would serve as useful background data in studies on RA monitoring strategies detecting subclinical PD synovitis. Thermographic temperatures were greater in patients with greater disease activity and can help discriminate ultrasound PD joint inflammation severity.
Collapse
Affiliation(s)
- York Kiat Tan
- Department of Rheumatology and Immunology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Rd, Singapore 169857, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| | - Julian Thumboo
- Department of Rheumatology and Immunology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
- Duke-NUS Medical School, 8 College Rd, Singapore 169857, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore 117597, Singapore
| |
Collapse
|
19
|
Ruan WJ, Xu SS, Xu DH, Li ZP. Orthopedic revolution: The emerging role of nanotechnology. World J Orthop 2024; 15:932-938. [DOI: 10.5312/wjo.v15.i10.932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/24/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024] Open
Abstract
This review summarizes the latest progress in orthopedic nanotechnology, explores innovative applications of nanofibers in tendon repair, and evaluates the potential of selenium and cerium oxide nanoparticles in osteoarthritis and osteoblast differentiation. This review also describes the emerging applications of injectable hydrogels in cartilage engineering, emphasizing the critical role of interdisciplinary research and highlighting the challenges and future prospects of integrating nanotechnology into orthopedic clinical practice. This comprehensive approach provides a holistic perspective on the transformative impact of nanotechnology in orthopedics, offering valuable insights for future research and clinical applications.
Collapse
Affiliation(s)
- Wen-Jie Ruan
- Department of Sports Medicine, Zhejiang Provincial People's Hospital (The Affiliated People's Hospital), Hangzhou 310000, Zhejiang Province, China
| | - Si-Si Xu
- School of Medicine, Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Dong-Hui Xu
- School of Medicine, Taizhou University, Taizhou 318000, Zhejiang Province, China
| | - Zhi-Peng Li
- The Second Department of Orthopedics, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, Henan Province, China
| |
Collapse
|
20
|
Tang L, Guo D, Jia D, Piao S, Fang C, Zhu Y, Wang Y, Pan Z. Exploring the therapeutic potential of "Tianyu" medicine pair in rheumatoid arthritis: an integrated study combining LC-MS/MS, bioinformatics, network pharmacology, and experimental validation. Front Med (Lausanne) 2024; 11:1475239. [PMID: 39430588 PMCID: PMC11488520 DOI: 10.3389/fmed.2024.1475239] [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: 08/03/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a widespread chronic autoimmune disease that primarily causes joint inflammation and damage. In advanced stages, RA can result in joint deformities and loss of function, severely impacting patients' quality of life. The "Tianyu" pair (TYP) is a traditional Chinese medicine formulation developed from clinical experience and has shown some effectiveness in treating RA. However, its role in the complex biological mechanisms underlying RA remains unclear and warrants further investigation. Methods We obtained gene sequencing data of synovial tissues from both RA patients and healthy individuals using two gene microarrays, GSE77298 and GSE55235, from the GEO database. Through an integrated approach involving bioinformatics, machine learning, and network pharmacology, we identified the core molecular targets of the "Tianyu" medicine pair (TYP) for RA treatment. Liquid chromatography-mass spectrometry was then employed to analyze the chemical components of TYP. To validate our findings, we conducted animal experiments with Wistar rats, comparing histopathological and key gene expression changes before and after TYP treatment. Results Our data analysis suggests that the onset of RA may be associated with inflammation-related immune cells involved in both adaptive and innate immune responses. Potential key targets for TYP treatment in RA include AKR1B10, MMP13, FABP4, NCF1, SPP1, COL1A1, and RASGRP1. Among the components of TYP, Kaempferol, Quercetin, and Salidroside were identified as key, with MMP13 and NCF1 showing the strongest binding affinity to these compounds. Animal experiments confirmed the findings from bioinformatics and network pharmacology, validating the key targets and therapeutic effects of TYP in treating RA. Conclusion Our study reveals that TYP has potential clinical value in the treatment of rheumatoid arthritis. This research enhances our understanding of RA's pathogenesis and provides insight into potential therapeutic mechanisms.
Collapse
Affiliation(s)
- Lu Tang
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Dingyuan Guo
- Department of Traditional Chinese Internal Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongye Jia
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Songlan Piao
- Department of Pathology Teaching and Research, Clinical Medical School, Changchun University of Chinese Medicine, Changchun, China
| | - Chunqiu Fang
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Yueya Zhu
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| | - Yinghang Wang
- Department of Rheumatology and Immunology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Zhi Pan
- Fangzheng Research Laboratory, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
21
|
Patil SJ, Thorat VM, Koparde AA, Bhinge SD, Chavan DD, Bhosale RR. Unlocking the Future: New Biologic Therapies for Rheumatoid Arthritis. Cureus 2024; 16:e72486. [PMID: 39600762 PMCID: PMC11592031 DOI: 10.7759/cureus.72486] [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: 10/13/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder that leads to joint destruction and functional disability. Traditional treatments, including disease-modifying antirheumatic drugs (DMARDs), often fail, leaving many patients without remission. The advent of biologic therapies that target specific immune system components (e.g., cytokines, T cells) has transformed RA treatment by offering new management options. These biologics (e.g., TNF inhibitors, interleukin blockers) are highly effective in controlling disease activity and preventing joint destruction. However, their use comes with safety concerns, particularly regarding immunosuppression and infection risks. Although still experimental, studies predict that future research will focus on enhancing the clinical response and safety of these agents through personalized approaches or novel mechanisms of action.
Collapse
Affiliation(s)
- Sarika J Patil
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Vandana M Thorat
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Akshada A Koparde
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Somnath D Bhinge
- Department of Pharmaceutical Chemistry, Rajarambapu College of Pharmacy, Kasegaon, IND
| | - Dhanashri D Chavan
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Rohit R Bhosale
- Department of Pharmaceutics, Krishna Foundation's Jaywant Institute of Pharmacy, Wathar, IND
| |
Collapse
|
22
|
Ahmed EA, Abdelsalam SA. Marine Bioactive Molecules as Inhibitors of the Janus Kinases: A Comparative Molecular Docking and Molecular Dynamics Simulation Approach. Curr Issues Mol Biol 2024; 46:10635-10650. [PMID: 39329982 PMCID: PMC11430628 DOI: 10.3390/cimb46090631] [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: 08/27/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
A treasure trove of naturally occurring biomolecules can be obtained from sea living organisms to be used as potential antioxidant and anti-inflammatory agents. These bioactive molecules can target signaling molecules involved in the severity of chronic autoimmune diseases such as rheumatoid arthritis (RA). The intracellular tyrosine kinases family, Janus kinases (JAKs, includes JAK1, JAK2, and JAK3), is implicated in the pathogenesis of RA through regulating several cytokines and inflammatory processes. In the present study, we conducted molecular docking and structural analysis investigations to explore the role of a set of bioactive molecules from marine sources that can be used as JAKs' specific inhibitors. Around 200 antioxidants and anti-inflammatory molecules out of thousands of marine molecules found at the Comprehensive Marine Natural Products Database (CMNPD) website, were used in that analysis. The details of the interacting residues were compared to the recent FDA approved inhibitors tofacitinib and baricitinib for data validation. The shortlisted critical amino acids residues of our pharmacophore-based virtual screening were LYS905, GLU957, LEU959, and ASP1003 at JAK1, GLU930 and LEU932 at JAK2, and GLU905 and CYS909 of JAK3. Interestingly, marine biomolecules such as Sargachromanol G, Isopseudopterosin E, Seco-Pseudopterosin, and CID 10071610 showed specific binding and significantly higher binding energy to JAK1 active/potential sites when being compared with the approved inhibitors. In addition, Zoanthoxanthin and Fuscoside E bind to JAK2's critical residues, GLU930 and LEU932. Moreover, Phorbaketal and Fuscoside E appear to be potential candidates that can inhibit JAK3 activity. These results were validated using molecular dynamics simulation for the docked complexes, JAK1(6sm8)/SG, JAK2 (3jy9)/ZAX, and JAK3 (6pjc)/Fuscoside E, where stable and lower binding energy were found based on analyzing set of parameters, discussed below (videos are attached). A promising role of these marine bioactive molecules can be confirmed in prospective preclinical/clinical investigations using rheumatoid arthritis models.
Collapse
Affiliation(s)
- Emad A. Ahmed
- Department of Biological Sciences, College of Science, King Faisal University, Hofouf 31982, Saudi Arabia
- Lab of Molecular Physiology, Zoology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt;
| | - Salah A. Abdelsalam
- Lab of Molecular Physiology, Zoology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt;
- Zoology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| |
Collapse
|
23
|
Swarnkar G, Naaz M, Mims D, Gupta P, Peterson T, Christopher MJ, Singamaneni S, Mbalaviele G, Abu-Amer Y. IĸBζ as a Central Modulator of Inflammatory Arthritis Pathogenesis. Arthritis Rheumatol 2024. [PMID: 39279148 DOI: 10.1002/art.42990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Current therapies targeting individual factors in inflammatory arthritis show variable efficacy, often requiring treatment with combinations of drugs, and are associated with undesirable side effects. NF-ĸB is critical for the production and function of most inflammatory cytokines. However, given its essential role in physiologic processes, targeting NF-ĸB is precarious. Hence, identifying pathways downstream of NF-ĸB that selectively govern the expression of inflammatory cytokines in inflammatory arthritis would be advantageous. We have previously identified IĸBζ as a unique inflammatory signature of NF-ĸB that controls the transcription of inflammatory cytokines only under pathologic conditions while sparing physiologic NF-ĸB signals. METHODS We generated mice harboring myeloid, lymphoid, and global deletion of Nfkbiz (the gene encoding IĸBζ). These models were subjected to serum transfer-induced arthritis. Additionally, pharmacologic inhibitors of IĸBζ were injected intraperitonially. Joint swelling, microcomputed tomography, immunohistochemistry, flow cytometry, and cytokine measurements were conducted using synovial tissue samples. RESULTS Global deletion of Nfkbiz or depletion of neutrophils (vastly IĸBζ+ cells) reduced inflammatory synovial cells and increased anti-inflammatory and regenerative synovial cells, plummeted expression of inflammatory factors and ameliorated experimental mouse inflammatory arthritis. Further, expression of immune responsive gene-1, the enzyme responsible for itaconate production, was increased in synovial cells. Accordingly, the itaconate derivative dimethyl itaconate (DI) inhibited IĸBζ-mediated inflammatory factors. Further, in silico screen identified 8-hydroxyquinoline (HQ) as a putative inhibitor of IĸBζ not affecting physiologic NF-ĸB activity. Congruently, systemic administration of either DI or HQ inhibited joint swelling and damage. CONCLUSION Our study positions IĸBζ as an inflammation-specific target for therapeutic consideration in rheumatoid arthritis because its inhibition spares the beneficial functions of NF-ĸB.
Collapse
Affiliation(s)
- Gaurav Swarnkar
- Washington University School of Medicine, St. Louis, Missouri
| | - Musarrat Naaz
- Washington University School of Medicine, St. Louis, Missouri
| | - Dorothy Mims
- Washington University School of Medicine, St. Louis, Missouri
| | | | - Timothy Peterson
- Washington University School of Medicine, HealthSpan Technologies, Inc., and Bioio, Inc., St. Louis, Missouri
| | | | | | | | - Yousef Abu-Amer
- Washington University School of Medicine and Shriners Hospital for Children, St. Louis, Missouri
| |
Collapse
|
24
|
Zhang K, Luo Z, Wang X. The association of common autoimmune diseases with autoimmune thyroiditis: a two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1383221. [PMID: 39314521 PMCID: PMC11416997 DOI: 10.3389/fendo.2024.1383221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/23/2024] [Indexed: 09/25/2024] Open
Abstract
Objective Numerous observational and retrospective studies have demonstrated an association between Autoimmune Thyroiditis (AIT) and various systemic Autoimmune Diseases (AIDs). However, the causal relationship between them remains uncertain. This study aims to investigate the causal link between AIT and diverse types of AIDs utilizing the Mendelian Randomization (MR) method. Method We assessed the causal relationship between AIT and eight prevalent AIDs. Summary statistics from genome-wide association studies (GWAS) were sourced from the FinnGen biobank and IEU Open GWAS database. Two-sample MR analyses were conducted, with the primary statistical approach being the Inverse Variance Weighting (IVW) method. This was complemented by a series of sensitivity analyses, and the robustness of the findings was evaluated through the estimation of heterogeneity and pleiotropy. Results When AIT was considered as the outcome, MR evidence suggested an association between Rheumatoid arthritis (RA), Type 1 diabetes (T1D), and Systemic lupus erythematosus (SLE) with AIT. Utilizing the Inverse Variance Weighting (IVW) method, we observed an increased risk of AIT with exposure to RA (P = 0.024, OR=1.25; 95% CI = 1.03, 1.52), T1D (P < 0.001, OR=1.27 95% CI = 1.11,1.46), and SLE (P = 0.037, OR=1.14; 95% CI = 1.04,1.26). Conversely, no significant genetic causal relationship with AIT was found for Sjögren's syndrome (SS), Ankylosing Spondylitis (AS), Multiple sclerosis (MS), Crohn's disease (CD), and Ulcerative colitis (UC). Conclusion This study identified RA, T1D, and SLE as triggering factors for AIT. The incidence rate of AIT in patients with RA, T1D, and SLE may be higher than that in the general population. Therefore, individuals with these three diseases should undergo regular monitoring of thyroid-related indicators.
Collapse
MESH Headings
- Humans
- Mendelian Randomization Analysis
- Thyroiditis, Autoimmune/genetics
- Thyroiditis, Autoimmune/epidemiology
- Thyroiditis, Autoimmune/complications
- Genome-Wide Association Study
- Autoimmune Diseases/genetics
- Autoimmune Diseases/epidemiology
- Autoimmune Diseases/complications
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/epidemiology
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/complications
- Arthritis, Rheumatoid/epidemiology
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/complications
- Lupus Erythematosus, Systemic/epidemiology
- Genetic Predisposition to Disease
- Polymorphism, Single Nucleotide
Collapse
Affiliation(s)
- Kaiyuan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Ziyue Luo
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xinchang Wang
- Department of Rheumatology, The Second Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
25
|
Qiu F, Xie D, Chen H, Wang Z, Huang J, Cao C, Liang Y, Yang X, He DY, Fu X, Lu A, Liang C. Generation of cytotoxic aptamers specifically targeting fibroblast-like synoviocytes by CSCT-SELEX for treatment of rheumatoid arthritis. Ann Rheum Dis 2024:ard-2024-225565. [PMID: 39237134 DOI: 10.1136/ard-2024-225565] [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: 01/22/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is an autoimmune disease characterised by aggressive fibroblast-like synoviocytes (FLSs). Very few RA patients-derived FLSs (RA-FLSs)-specific surface signatures have been identified, and there is currently no approved targeted therapy for RA-FLSs. This study aimed to screen therapeutic aptamers with cell-targeting and cytotoxic properties against RA-FLSs and to uncover the molecular targets and mechanism of action of the screened aptamers. METHODS We developed a cell-specific and cytotoxic systematic evolution of ligands by exponential enrichment (CSCT-SELEX) method to screen the therapeutic aptamers without prior knowledge of the surface signatures of RA-FLSs. The molecular targets and mechanisms of action of the screened aptamers were determined by pull-down assays and RNA sequencing. The therapeutic efficacy of the screened aptamers was examined in arthritic mouse models. RESULTS We obtained an aptamer SAPT8 that selectively recognised and killed RA-FLSs. The molecular target of SAPT8 was nucleolin (NCL), a shuttling protein overexpressed on the surface and involved in the tumor-like transformation of RA-FLSs. Mechanistically, SAPT8 interacted with the surface NCL and was internalised to achieve lysosomal degradation of NCL, leading to the upregulation of proapoptotic p53 and downregulation of antiapoptotic B-cell lymphoma 2 (Bcl-2) in RA-FLSs. When administrated systemically to arthritic mice, SAPT8 accumulated in the inflamed FLSs of joints. SAPT8 monotherapy or its combination with tumour necrosis factor (TNF)-targeted biologics was shown to relieve arthritis in mouse models. CONCLUSIONS CSCT-SELEX could be a promising strategy for developing cell-targeting and cytotoxic aptamers. SAPT8 aptamer selectively ablates RA-FLSs via modulating NCL-p53/Bcl-2 signalling, representing a potential alternative or complementary therapy for RA.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | | | - Xu Yang
- Department of Computational Biology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong-Yi He
- Department of Rheumatology, Shanghai Guanghua Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
26
|
Chang J, Wang G. A retrospective study of efficacy of tofacitinib combined with bDMARDs in the treatment of rheumatoid arthritis patients with inadequate response to bDMARDs. Int J Rheum Dis 2024; 27:e15311. [PMID: 39198040 DOI: 10.1111/1756-185x.15311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/12/2024] [Accepted: 08/15/2024] [Indexed: 09/01/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation, joint swelling, and pain involving multiple joints. While biologic disease-modifying antirheumatic drugs (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) are popular treatments for RA, there is limited research on their combined use. This study examined a cohort of RA patients who demonstrated inadequate response to bDMARDs and subsequently initiated combination therapy with tofacitinib and bDMARDs, assessing both the efficacy and safety profile of this therapeutic approach. METHODS In this study, we retrospectively collected the electronic medical records (EMR) of 62 adult patients with RA who were admitted to the Fourth Affiliated Hospital Zhejiang University School of Medicine between August 2018 and December 2022. All patients had received at least one bDMARD treatment for more than 3 months and still exhibited moderate-to-high disease activity. Tofacitinib 5 mg bid was added to their original biological treatment in 28 cases, and other 34 cases switched to another bDMARD or tsDMARD as control group. Treatment was continued for 24 weeks following the initiation of combination therapy. Changes in DAS28-ESR and ACR20, 50, 70 response rates at week 24 were collected and analyzed from baseline, while changes in C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) at weeks 4, 8, 12, 24 were also collected and analyzed. RESULTS After 24 weeks of treatment, the DAS28-ESR score in combined treatment group decreased significantly from a baseline of 5.26 ± 0.90 (3.87-8.31) to 2.67 ± 0.86 (1.41-5.11), with remission achieved by 19 patients (67.9%) and low disease activity achieved by five patients (17.9%). The DAS28-ESR in the control group exhibited a decrease from 5.20 ± 0.77 (3.87-7.23) at baseline to 3.25 ± 1.29 (1.54-5.69). In all, 13 patients (38.2%) achieved remission, while another 11 patients (32.4%) achieved low disease activity. The ACR20, 50, 70 response rates were 85.71%, 75%, and 39.29% in the combined treatment group, whereas it were 75.0%, 53.57%, 21.43% in the control group. Additionally, both ESR and CRP levels decreased significantly during the course of treatment without any reported adverse events leading to discontinuation. CONCLUSION Our findings offer some evidence, supporting the effectiveness and safety of combining bDMARD with JAKi tofacitinib in RA patients who have an inadequate response to bDMARD monotherapy. This combination effectively manages disease activity while maintaining a relatively low and manageable incidence of adverse events. Further prospective randomized controlled trials with large sample sizes are anticipated to provide evidence-based medical support.
Collapse
Affiliation(s)
- Jie Chang
- Department of Rheumatology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Gang Wang
- Department of Rheumatology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| |
Collapse
|
27
|
Guan Y, Li X, Yang H, Xu S, Shi L, Liu Y, Kong L, Qin Y. Role and mechanism of IRF9 in promoting the progression of rheumatoid arthritis by regulating macrophage polarization via PSMA5. Heliyon 2024; 10:e35589. [PMID: 39170377 PMCID: PMC11336755 DOI: 10.1016/j.heliyon.2024.e35589] [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: 04/02/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Aim To explore the mechanisms of IRF9 in the progression of rheumatoid arthritis(RA), and the effects of IRF9 on M1/M2 polarization. Methods RA dataset (GSE55457) was downloaded from GEO. Correlation analysis between IRF9 and its downstream target protein PSMA5 was performed using bioinformatics analysis. The M1/M2 cell ratio of peripheral blood mononuclear cells which from 20 healthy specimen and 40 RA patients was determined. The expression of IRF9 and PSMA5 was detected using qPCR and Western blot. Then, knockdown IRF9 in RAW264.7 cell line (sh-IRF9 RAW264.7) was constructed. The effect of sh-IRF9 RAW264.7 on RA was explored by constructing a CIA mouse model. Results IRF9 is upregulated in RA and is of good early screening effect. The results of pathway analysis showed that IRF9 targets and regulates the PSMA5 signaling pathway. IRF9 and PSMA5 were significantly elevated in RA patients, M1/M2 ratio was also increased. The effects of IRF9 on RAW264.7 macrophages were deeply explored in vitro, revealing that knockdown of IRF9 suppressed PSMA5, M1/M2 ratio and the secretion of pro-inflammatory factor in RAW264.7. In mouse in vivo experiments, sh-IRF9 RAW264.7 cells were found to modulate RA by downregulating PSMA5, modulating the M1/M2 ratio through enhancing the anti-inflammatory factor, and suppressing the pro-inflammatory factor. Conclusion IRF9 promoted the progression of RA via regulating macrophage polarization through PSMA5.
Collapse
Affiliation(s)
- Yue Guan
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Xin Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Hemin Yang
- Central Laboratory, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Siyu Xu
- Inspection Center, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Lidong Shi
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Yangyang Liu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Lingdan Kong
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Ying Qin
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| |
Collapse
|
28
|
Liu Q, Wang J, Ding C, Chu Y, Jiang F, Hu Y, Li H, Wang Q. Sinomenine Alleviates Rheumatoid Arthritis by Suppressing the PI3K-Akt Signaling Pathway, as Demonstrated Through Network Pharmacology, Molecular Docking, and Experimental Validation. Drug Des Devel Ther 2024; 18:3523-3545. [PMID: 39135759 PMCID: PMC11317229 DOI: 10.2147/dddt.s475959] [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: 05/22/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
Purpose Sinomenine (SIN) is commonly used in Traditional Chinese Medicine (TCM) as a respected remedy for rheumatoid arthritis (RA). Nevertheless, the therapeutic mechanism of SIN in RA remains incompletely understood. This study aimed to delve into the molecular mechanism of SIN in the treatment of RA. Methods The potential targets of SIN were predicted using the TCMSP server, STITCH database, and SwissTarget Prediction. Differentially expressed genes (DEGs) in RA were obtained from the GEO database. Enrichment analyses and molecular docking were conducted to explore the potential mechanism of SIN in the treatment of RA. In vitro and in vivo studies were conducted to validate the intervention effects of SIN on rheumatoid arthritis, as determined through network pharmacology analyses. Results A total of 39 potential targets associated with the therapeutic effects of SIN in RA were identified. Enrichment analysis revealed that these potential targets are primarily enriched in PI3K-Akt signaling pathway, and the molecular docking suggests that SIN may act on specific proteins in the pathway. Experimental results have shown that exposure to SIN inhibits cytokine secretion, promotes apoptosis, reduces metastasis and invasion, and blocks the activation of the PI3K-Akt signaling pathway in RA fibroblast-like synoviocytes (RA-FLS). Moreover, SIN treatment alleviated arthritis-related symptoms and regulated the differentiation of CD4+ T cells in the spleen of collagen-induced arthritis (CIA) mice. Conclusion By utilizing network pharmacology, molecular modeling, and in vitro/in vivo validation, this study demonstrates that SIN can alleviate RA by inhibiting the PI3K-Akt signaling pathway. These findings enhance the understanding of the therapeutic mechanisms of SIN in RA, offering a stronger theoretical foundation for its future clinical application.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Jian Wang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Chunhui Ding
- Department of Pharmacy, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Ying Chu
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Fengying Jiang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Yunxia Hu
- Department of Rheumatology and Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Haifeng Li
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214000, People's Republic of China
| |
Collapse
|
29
|
Pelissier A, Laragione T, Gulko PS, Rodríguez Martínez M. Cell-specific gene networks and drivers in rheumatoid arthritis synovial tissues. Front Immunol 2024; 15:1428773. [PMID: 39161769 PMCID: PMC11330812 DOI: 10.3389/fimmu.2024.1428773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/24/2024] [Indexed: 08/21/2024] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune and inflammatory disease characterized by inflammation and hyperplasia of the synovial tissues. RA pathogenesis involves multiple cell types, genes, transcription factors (TFs) and networks. Yet, little is known about the TFs, and key drivers and networks regulating cell function and disease at the synovial tissue level, which is the site of disease. In the present study, we used available RNA-seq databases generated from synovial tissues and developed a novel approach to elucidate cell type-specific regulatory networks on synovial tissue genes in RA. We leverage established computational methodologies to infer sample-specific gene regulatory networks and applied statistical methods to compare network properties across phenotypic groups (RA versus osteoarthritis). We developed computational approaches to rank TFs based on their contribution to the observed phenotypic differences between RA and controls across different cell types. We identified 18 (fibroblast-like synoviocyte), 16 (T cells), 19 (B cells) and 11 (monocyte) key regulators in RA synovial tissues. Interestingly, fibroblast-like synoviocyte (FLS) and B cells were driven by multiple independent co-regulatory TF clusters that included MITF, HLX, BACH1 (FLS) and KLF13, FOSB, FOSL1 (B cells). However, monocytes were collectively governed by a single cluster of TF drivers, responsible for the main phenotypic differences between RA and controls, which included RFX5, IRF9, CREB5. Among several cell subset and pathway changes, we also detected reduced presence of Natural killer T (NKT) cells and eosinophils in RA synovial tissues. Overall, our novel approach identified new and previously unsuspected Key driver genes (KDG), TF and networks and should help better understanding individual cell regulation and co-regulatory networks in RA pathogenesis, as well as potentially generate new targets for treatment.
Collapse
Affiliation(s)
- Aurelien Pelissier
- Institute of Computational Life Sciences, Zürich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
- AI for Scientific Discovery, IBM Research Europe, Rüschlikon, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Teresina Laragione
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Percio S. Gulko
- Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - María Rodríguez Martínez
- AI for Scientific Discovery, IBM Research Europe, Rüschlikon, Switzerland
- Department of Biomedical Informatics & Data Science, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
30
|
Su X, Wang X, Zhang X, Sun Y, Jia Y. β-Indole-3-acetic acid attenuated collagen-induced arthritis through reducing the ubiquitination of Foxp3 via the AhR-TAZ-Tip60 pathway. Immunol Res 2024; 72:741-753. [PMID: 38630408 DOI: 10.1007/s12026-024-09480-x] [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: 02/09/2024] [Accepted: 04/09/2024] [Indexed: 08/28/2024]
Abstract
Massive evidence shows that intestinal tryptophan metabolites affected by intestinal flora can modulate the progression of rheumatoid arthritis (RA). However, the effects and mechanisms of intestinal tryptophan metabolites on RA are not yet detailed. Herein, we investigated the protective effects of intestinal tryptophan metabolites on RA and its detailed mechanisms. In this study, the collagen-induced arthritis (CIA) rat model was established. Based on metabolomics analysis, the contents of β-indole-3-acetic acid (IAA), indolylpropionic acid, and indole-3-β-acrylic acid in the sera of CIA rats were significantly less compared with those of the normal rats. Under the condition of Treg or Th17 cell differentiation, IAA significantly promoted the differentiation and activation of Treg cells instead of Th17 cells. Intestinal tryptophan metabolites are well-known endogenic ligands of aryl hydrocarbon receptor (AhR). Not surprisingly, IAA increased the level of Foxp3 through activating the AhR pathway. Interestingly, IAA had little impact on the level of Foxp3 mRNA, but reducing the ubiquitination and degradation of Foxp3. Mechanically, IAA reduced the expression of the transcriptional coactivator TAZ, which was almost completely reversed by either AhR antagonist CH223191 or siRNA. In vitro, IAA decreased the combination of TAZ and the histone acetyltransferase Tip60, while it increased the combination of Tip60 and Foxp3. In CIA rats, oral administration of IAA increased the number of Treg cells and relieved the inflammation. A combined use with CH223191 almost abolished the effect of IAA. Taken together, IAA attenuated CIA by promoting the differentiation of Treg cells through reducing the ubiquitination of Foxp3 via the AhR-TAZ-Tip60 pathway.
Collapse
Affiliation(s)
- Xiaoran Su
- Department of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Xinliu Wang
- Department of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Xin Zhang
- Department of Integrative Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yajie Sun
- Department of Chinese Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yugai Jia
- Department of Pharmacology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China.
- Hebei International Cooperation Center for Ion Channel Function and Innovative Traditional Chinese Medicine, Shijiazhuang, 050091, China.
| |
Collapse
|
31
|
Roghani SA, Shamsi A, Jalili C, Jalili F, Lotfi R, Garman N, Rostampour R, Taghadosi M. Interleukin-6 positively correlates with cardiovascular disease predictor algorithms and biomarker in rheumatoid arthritis patients. J Cell Mol Med 2024; 28:e70028. [PMID: 39160453 PMCID: PMC11333199 DOI: 10.1111/jcmm.70028] [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: 02/07/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024] Open
Abstract
Chronic inflammation is believed as the main culprit of the link between cardiovascular disease (CVD) and rheumatoid arthritis (RA). Interleukin-6 (IL-6) is a pro-inflammatory cytokine with a key role in RA pathophysiology and also correlates with joint destruction and disease activity. This study evaluates the association between IL-6 plasma level and cardiac biomarker NT-proBNP, HS-CRP, CVD predictor algorithms, Framingham Risk Score (FRS) and Systematic Coronary Risk Evaluation (SCORE), as well as with CXCL9 and its receptor, CXCR3 in RA patients compared to the controls. Sixty RA patients (30 early and 30 late) and 30 healthy persons were included in this study. IL-6 and NT-proBNP plasma levels were measured by the ELISA. Also, HS-CRP plasma levels were quantified using the immunoturbidimetric assay. The CVD risk was assessed by the FRS and SCORE. IL-6 plasma levels were significantly higher in the early and late RA patients compared to the controls (p < 0.001). There was a positive correlation between IL-6 with DAS-28 (p = 0.007, r = 0.346), BPS (p = 0.002, r = 0.396), BPD (p = 0.046, r = 0.259), SCORE (p < 0.001, r = 0.472), and FRS (p < 0.001, r = 0.553), and a negative association with HDL (p = 0.037, r = -0.270), in the patients. Also, IL-6 plasma level positively correlated with HS-CRP (p = 0.021, r = 0.297) and NT-proBNP (p = 0.045, r = 0.260) in the patients. Furthermore, a positive association was found between IL-6 plasma levels and CXCL9 (p = 0.002, r = 0.386), and CXCR3 (p = 0.018, r = 0.304) in the patients. Given the interesting association between IL-6 with various variables of CVD, IL-6 may be considered a biomarker for assessing the risk for future cardiovascular events in RA patients.
Collapse
Affiliation(s)
- Seyed Askar Roghani
- Immunology Department, Faculty of MedicineKermanshah University of Medical SciencesKermanshahIran
- Clinical Research Development Center, Imam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
- Medical Biology Research Center, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Afsaneh Shamsi
- Immunology Department, Faculty of MedicineKermanshah University of Medical SciencesKermanshahIran
| | - Cyrus Jalili
- Medical Biology Research Center, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| | - Farnaz Jalili
- University of AdelaideAdelaideSouth AustraliaAustralia
| | - Ramin Lotfi
- Blood Transfusion Research CenterHigh Institute for Research and Education in Transfusion MedicineTehranIran
- Clinical Research Development Center, Tohid HospitalKurdistan University of Medical SciencesSanandajIran
| | - Nima Garman
- Department of Biology, Faculty of Basic SciencesUniversity of GuilanRashtIran
| | - Rezvan Rostampour
- Clinical Research Development Center, Imam Reza HospitalKermanshah University of Medical SciencesKermanshahIran
| | - Mahdi Taghadosi
- Cardiovascular Research Center, Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
32
|
Li Y, Zhang Z, Liu C, Kang Z, Li Z. Significance of High-Frequency Sonography for the Subclinical Progression of Rheumatoid Arthritis. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2024; 43:1509-1520. [PMID: 38720412 DOI: 10.1002/jum.16474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 07/14/2024]
Abstract
OBJECTIVES This study aims to investigate the use of high-frequency sonography as a tool for detecting inflammatory and destructive changes in the hand and foot joints of patients with early and long-term RA. METHODS This study employs a prospective cohort design involving 162 patients diagnosed with Rheumatoid arthritis (RA) who meet the American College of Rheumatology/European League Against Rheumatism (ACR/EULAR) classification criteria. Patients were divided into two groups based on disease duration: Group 1 (n = 74) included patients with a disease duration of up to 2 years, or early РА (ERA;), Group 2 (n = 88) consisted of patients with a disease duration exceeding 2 years, or long-term persistent РА (LtRA). All patients underwent a clinical assessment of their joints, as well as radiography and arthrosonography, at the beginning of the study and again at 6 and 12 months later. RESULTS In the general group of patients, ultrasound examination revealed signs of synovitis in the joints of the hands more frequently (66%) compared with clinical examination (56% by a number of swollen joints [NSJ] and 55% by a number of painful joints [NPJ], P < .01). After 6 months of treatment, 12% of the patients achieved full US remission and 24% achieved partial US remission. CONCLUSIONS Within the scope of comprehensive RA diagnostics, arthrosonography of the joints of the hands and feet, utilizing a combination of greyscale and power Doppler, may surpass radiography in detecting early RA. This method allows for a more accurate assessment of disease activity and progression rates.
Collapse
Affiliation(s)
- Yudan Li
- Ultrasonography Division, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Ziteng Zhang
- Hepato-Biliary Surgery Division, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Cuicui Liu
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhilong Kang
- Hepato-Biliary Surgery Division, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Zhiyan Li
- Ultrasonography Division, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
33
|
Jin Y, Chang C, Zhou X, Zhang R, Jiang P, Wei K, Xu L, Shi Y, Yang G, Lv X, Zheng Y, He D. LncRNA NONHSAT042241 inhibits rheumatoid synovial proliferation, inflammation and aggression via inactivating WNT/β-catenin signaling pathway. Autoimmunity 2024; 57:2387076. [PMID: 39229919 DOI: 10.1080/08916934.2024.2387076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/08/2024] [Accepted: 07/27/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE This study aims to explore the effect of NONHSAT042241 on the function of rheumatoid arthritis -fibroblast-like synoviocyte (RA-FLS) and the underlying mechanisms. METHODS RA-FLS was treated with NONHSAT042241 overexpression and NONHSAT042241 knockdown lentiviruses. Cell counting kit-8 (CCK-8) assay, colony formation assay, flow cytometry, Transwell assay, western-blot, ELISA, and qRT-PCR were used to measure the changes of cell proliferation, apoptosis, invasion, secretion of inflammatory cytokines and matrix metalloproteinases (MMPs). Fluorescent in situ hybridization (FISH) assay, RNA pull-down assay, mass spectrometry (MS) and RNA immunoprecipitation (RIP) were used to find the target proteins that bond to NONHSAT042241, and western-blot was used to detect the expression of related proteins of Wnt/β-catenin signaling pathway. RESULTS Overexpression of NONHSAT042241 inhibited the proliferation of RA-FLS (p < 0.05), invasion, secretion of pro-inflammatory factors (IL-1and IL-6) and MMPs (MMP-1 and MMP-3) (p < 0.05), and elevated the level of pro-apoptotic factors (Bax and cleaved caspase3), while NONHSAT042241 knockdown had the opposite effect. NONHSAT042241 can directly bind to hnRNP D, and down-regulated the expression of β-catenin (p < 0.05), p-GSK-3β (p < 0.05), Cyclin D1 (p < 0.05), PCNA (p < 0.05), and thus reduced the cell proliferation. CONCLUSION NONHSAT042241 may inhibit FLS-mediated rheumatoid synovial proliferation, inflammation and aggression. The underlying mechanisms may be that NONHSAT042241 inhibits the activity of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yehua Jin
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Cen Chang
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Xinpeng Zhou
- Department of Rheumatology, Shandong Province Hospital of Traditional Chinese Medicine, Jinan, China
| | - Runrun Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ping Jiang
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai Wei
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linshuai Xu
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Shi
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guizhen Yang
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinliang Lv
- Department of Rheumatology, Traditional Chinese Medicine Hospital of Inner Mongolia Autonomous Region, Hohhot, Inner Mongolia Autonomous Region, China
| | - Yuejuan Zheng
- Center for Traditional Chinese Medicine and Immunology Research, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongyi He
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- Department of Rheumatology, Shandong Province Hospital of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
34
|
Arturo NVC, Ivan GNJ, Betsabe CH, Emilio PGE, Yussef EG, Alejandra RJN, Tonatiuh GH, Alejandra VV, Ismael NA, Elena TSS, Manuel PGJ, Heriberto JC, Gerardo AAE, Laura GL, Miriam SCA. Genetic Variants of the Receptor Activator Nuclear of κB Ligand Gene Increase the Risk of Rheumatoid Arthritis in a Mexican Mestizo Population: A Case-Control Study. Genes (Basel) 2024; 15:907. [PMID: 39062686 PMCID: PMC11275863 DOI: 10.3390/genes15070907] [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: 06/20/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
The Receptor Activator Nuclear of κB Ligand (RANKL) plays an important function in immune responses, activating osteoclast cells and unchanged bone resorption, which in turn leads to bone erosion and inflammation. Genetic variants in the promoter region of the RANKL gene could lead to a higher risk of rheumatoid arthritis (RA). OBJECTIVE To assess the association of rs9533155 (-693C>G) and rs9533156 (-643T>C) genetic variants with RA risk. METHODS A case-control study was carried out. A total of 94 patients with RA (RA group) and 134 subjects without any rheumatologic disease (control group) were included. Genetic DNA was extracted from peripheral white blood cells (leukocytes). Genetic variant rs9533155 (-693C>G) was screened by an approach based on Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP), while rs9533156 (-643T>C) was screened using quantitative polymerase chain reaction (qPCR) with TaqMan probes. RANKL serum levels were measured by ELISA. RESULTS For rs9533155 (-693C>G), the polymorphic homozygous genotype frequencies (CC) were higher in the RA group (p = 0.006). Individuals carrying the risk genotype presented higher levels of serum RANKL. Carriers of the polymorphic homozygous genotype in the dominant model (CC vs. CG + GG) had an increased risk of developing RA (OR: 1.8, 95% CI 1.04 to 3.1). No association between rs9533156 (-643T>C) and the haplotypes with RA risk was observed. CONCLUSION The rs9533155 (-693C>G) genetic variant exhibits a potential role in RA risk. The studied population had no association with the rs9533156 (-643T>C) genetic variant.
Collapse
Affiliation(s)
- Nava-Valdivia Cesar Arturo
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Gamez-Nava Jorge Ivan
- Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Contreras-Haro Betsabe
- Unidad de Investigación Biomédica 02, UMAE, Hospital de Especialidades, Centro Médico Nacional de Occidente, IMSS, Guadalajara 44349, Mexico
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Guadalajara 45400, Mexico
| | - Perez-Guerrero Edsaul Emilio
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Esparza-Guerrero Yussef
- Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Rodriguez-Jimenez Norma Alejandra
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Gonzalez-Heredia Tonatiuh
- Unidad de Investigación Biomédica 02, UMAE, Hospital de Especialidades, Centro Médico Nacional de Occidente, IMSS, Guadalajara 44349, Mexico
| | - Villagomez-Vega Alejandra
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Guadalajara 45400, Mexico
| | - Nuño-Arana Ismael
- Departamento de Ciencias Biomédicas, Centro Universitario de Tonalá, Universidad de Guadalajara, Guadalajara 45400, Mexico
| | - Totsuka-Sutto Sylvia Elena
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Ponce-Guarneros Juan Manuel
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jacobo-Cuevas Heriberto
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alvarez-Ayala Efren Gerardo
- Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Gonzalez-Lopez Laura
- Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Saldaña-Cruz Ana Miriam
- Instituto de Terapéutica Experimental y Clínica, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| |
Collapse
|
35
|
Guo C, Peng J, Cheng P, Yang C, Gong S, Zhang L, Zhang T, Peng J. Mechanistic elucidation of ferroptosis and ferritinophagy: implications for advancing our understanding of arthritis. Front Physiol 2024; 15:1290234. [PMID: 39022306 PMCID: PMC11251907 DOI: 10.3389/fphys.2024.1290234] [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: 09/07/2023] [Accepted: 02/23/2024] [Indexed: 07/20/2024] Open
Abstract
In recent years, the emerging phenomenon of ferroptosis has garnered significant attention as a distinctive mode of programmed cell death. Distinguished by its reliance on iron and dependence on reactive oxygen species (ROS), ferroptosis has emerged as a subject of extensive investigation. Mechanistically, this intricate process involves perturbations in iron homeostasis, dampening of system Xc-activity, morphological dynamics within mitochondria, and the onset of lipid peroxidation. Additionally, the concomitant phenomenon of ferritinophagy, the autophagic degradation of ferritin, assumes a pivotal role by facilitating the liberation of iron ions from ferritin, thereby advancing the progression of ferroptosis. This discussion thoroughly examines the detailed cell structures and basic processes behind ferroptosis and ferritinophagy. Moreover, it scrutinizes the intricate web of regulators that orchestrate these processes and examines their intricate interplay within the context of joint disorders. Against the backdrop of an annual increase in cases of osteoarthritis, rheumatoid arthritis, and gout, these narrative sheds light on the intriguing crossroads of pathophysiology by dissecting the intricate interrelationships between joint diseases, ferroptosis, and ferritinophagy. The newfound insights contribute fresh perspectives and promising therapeutic avenues, potentially revolutionizing the landscape of joint disease management.
Collapse
Affiliation(s)
- Caopei Guo
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Jiaze Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Piaotao Cheng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Chengbing Yang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Shouhang Gong
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Lin Zhang
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiachen Peng
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Joint Orthopaedic Research Center of Zunyi Medical University, University of Rochester Medical Center, Zunyi, China
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi, China
| |
Collapse
|
36
|
Wu YZ, Chen WY, Zeng Y, Ji QL, Yang Y, Guo XL, Wang X. Inflammation-Responsive Mesoporous Silica Nanoparticles with Synergistic Anti-inflammatory and Joint Protection Effects for Rheumatoid Arthritis Treatment. Pharm Res 2024; 41:1493-1505. [PMID: 38918308 DOI: 10.1007/s11095-024-03732-z] [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: 04/05/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE Joint destruction is a major burden and an unsolved problem in rheumatoid arthritis (RA) patients. We designed an intra-articular mesoporous silica nanosystem (MSN-TP@PDA-GlcN) with anti-inflammatory and joint protection effects. The nanosystem was synthesized by encapsulating triptolide (TP) in mesoporous silica nanoparticles and coating it with pH-sensitive polydopamine (PDA) and glucosamine (GlcN) grafting on the PDA. The nano-drug delivery system with anti-inflammatory and joint protection effects should have good potency against RA. METHODS A template method was used to synthesize mesoporous silica (MSN). MSN-TP@PDA-GlcN was synthesized via MSN loading with TP, coating with PDA and grafting of GlcN on PDA. The drug release behavior was tested. A cellular inflammatory model and a rat RA model were used to evaluate the effects on RA. In vivo imaging and microdialysis (MD) system were used to analyze the sustained release and pharmacokinetics in RA rats. RESULTS TMSN-TP@PDA-GlcN was stable, had good biocompatibility, and exhibited sustained release of drugs in acidic environments. It had excellent anti-inflammatory effects in vitro and in vivo. It also effectively repaired joint destruction in vivo without causing any tissue toxicity. In vivo imaging and pharmacokinetics experiments showed that the nanosystem prolonged the residence time, lowered the Cmax value and enhanced the relative bioavailability of TP. CONCLUSIONS These results demonstrated that MSN-TP@PDA-GlcN sustained the release of drugs in inflammatory joints and produced effective anti-inflammatory and joint protection effects on RA. This study provides a new strategy for the treatment of RA.
Collapse
Affiliation(s)
- Ye-Zhen Wu
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China
| | - Wen-Yu Chen
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China
| | - Ying Zeng
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China
| | - Qi-Lin Ji
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China
| | - Yue Yang
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China
| | - Xu-Liang Guo
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China.
| | - Xiu Wang
- The Faculty of Pharmacy, Bengbu Medical University, Bengbu Anhui, 233030, P. R. China.
| |
Collapse
|
37
|
Liu D, Fu Q, Liu LG, Li W, Qi F, Liu J, Shang L, Wang X, Yang F, Li J, Lu D, Feng H, Zhang Z, Chen Y, Liang J, Yao J, Lv H, Li R, Wang J, Wu D, Liu Y, Xia C, Li W. Screening of potentially active compounds against rheumatoid arthritis in the Juan-Bi decoction using systems pharmacology and animal experiments. Front Cell Dev Biol 2024; 12:1396890. [PMID: 38983788 PMCID: PMC11231194 DOI: 10.3389/fcell.2024.1396890] [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: 03/06/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Background: The Juan-Bi decoction (JBD) is a classic traditional Chinese medicines (TCMs) prescription for the treatment of rheumatoid arthritis (RA). However, the active compounds of the JBD in RA treatment remain unclear. Aim: The aim of this study is to screen effective compounds in the JBD for RA treatment using systems pharmacology and experimental approaches. Method: Botanical drugs and compounds in the JBD were acquired from multiple public TCM databases. All compounds were initially screened using absorption, distribution, metabolism, excretion, and toxicity (ADMET) and physicochemical properties, and then a target prediction was performed. RA pathological genes were acquired from the DisGeNet database. Potential active compounds were screened by constructing a compound-target-pathogenic gene (C-T-P) network and calculating the cumulative interaction intensity of the compounds on pathogenic genes. The effectiveness of the compounds was verified using lipopolysaccharide (LPS)-induced RAW.264.7 cells and collagen-induced arthritis (CIA) mouse models. Results: We screened 15 potentially active compounds in the JBD for RA treatment. These compounds primarily act on multiple metabolic pathways, immune pathways, and signaling transduction pathways. Furthermore, in vivo and in vitro experiments showed that bornyl acetate (BAC) alleviated joint damage, and inflammatory cells infiltrated and facilitated a smooth cartilage surface via the suppression of the steroid hormone biosynthesis. Conclusion: We screened potential compounds in the JBD for the treatment of RA using systems pharmacology approaches. In particular, BAC had an anti-rheumatic effect, and future studies are required to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Dahai Liu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Qiang Fu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Leyna G Liu
- College of Letters and Science, University of California, Berkeley, Berkeley, CA, United States
| | - Wenwen Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fei Qi
- School of Clinical Medicine, Anhui Medical College, Hefei, Anhui, China
| | - Justin Liu
- Department of Statistics, University of California, Riverside, Riverside, CA, United States
| | - Lifeng Shang
- Guangdong Andao Medical Apparatus and Instruments Co., Ltd, Foshan, Guangdong, China
| | - Xiu Wang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Fang Yang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jie Li
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Daoqiang Lu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Huiying Feng
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Ziwen Zhang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Yiqing Chen
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Junru Liang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jiayi Yao
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Hua Lv
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Riwang Li
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jun Wang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Di Wu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Yuxi Liu
- Department of Traditional Chinese Medicine and Gynecology, Shunde Women and Children's Hospital of Guangdong Medical University (Maternity and Child Healthcare Hospital of Shunde Foshan), Foshan, Guangdong, China
| | - Chenglai Xia
- Foshan Maternity and Child Healthcare Hospital, Foshan, China
| | - Wenxing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Luo J, Zhu Y, Yu Y, Chen Y, He K, Liu J. Sinomenine treats rheumatoid arthritis by inhibiting MMP9 and inflammatory cytokines expression: bioinformatics analysis and experimental validation. Sci Rep 2024; 14:12786. [PMID: 38834626 PMCID: PMC11151427 DOI: 10.1038/s41598-024-61769-x] [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: 11/16/2023] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease marked by inflammatory cell infiltration and joint damage. The Chinese government has approved the prescription medication sinomenine (SIN), an effective anti-inflammation drug, for treating RA. This study evaluated the possible anti-inflammatory actions of SIN in RA based on bioinformatics analysis and experiments. Six microarray datasets were acquired from the gene expression omnibus (GEO) database. We used R software to identify differentially expressed genes (DEGs) and perform function evaluations. The CIBERSORT was used to calculate the abundance of 22 infiltrating immune cells. The weighted gene co-expression network analysis (WGCNA) was used to discover genes associated with M1 macrophages. Four public datasets were used to predict the genes of SIN. Following that, function enrichment analysis for hub genes was performed. The cytoHubba and least absolute shrinkage and selection operator (LASSO) were employed to select hub genes, and their diagnostic effectiveness was predicted using the receiver operator characteristic (ROC) curve. Molecular docking was undertaken to confirm the affinity between the SIN and hub gene. Furthermore, the therapeutic efficacy of SIN was validated in LPS-induced RAW264.7 cells line using Western blot and Enzyme-linked immunosorbent assay (ELISA). The matrix metalloproteinase 9 (MMP9) was identified as the hub M1 macrophages-related biomarker in RA using bioinformatic analysis and molecular docking. Our study indicated that MMP9 took part in IL-17 and TNF signaling pathways. Furthermore, we found that SIN suppresses the MMP9 protein overexpression and pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the LPS-induced RAW264.7 cell line. In conclusion, our work sheds new light on the pathophysiology of RA and identifies MMP9 as a possible RA key gene. In conclusion, the above findings demonstrate that SIN, from an emerging research perspective, might be a potential cost-effective anti-inflammatory medication for treating RA.
Collapse
Affiliation(s)
- Jinfang Luo
- Department of Basic Medicine, Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, People's Republic of China
| | - Yi Zhu
- Department of Basic Medicine, Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, People's Republic of China
| | - Yang Yu
- Department of Basic Medicine, Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, People's Republic of China
| | - Yujie Chen
- College of Clinical Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116622, People's Republic of China
| | - Kang He
- Department of Basic Medicine, Department of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, People's Republic of China.
| | - Jianxin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Jinxi South Road, Huaihua, 418000, People's Republic of China.
| |
Collapse
|
39
|
Piplani S, Jelic V, Johnson A, Shah U, Kolli S, Kong S, Tanasijevic N, Bejugam VR, Goguri SR, Mogga P, Kasire SP, Chaturvedi S, Jain P. Prevalence, Causes and Outcomes of Acute Gastrointestinal Bleeding in Rheumatoid Arthritis: A Systematic Review and Meta-Analysis. Mediterr J Rheumatol 2024; 35:210-219. [PMID: 39211013 PMCID: PMC11350423 DOI: 10.31138/mjr.230324.pca] [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: 03/23/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 09/04/2024] Open
Abstract
Aim The present study aims to investigate the prevalence, causes and outcomes of acute gastrointestinal (GI) bleeding in Rheumatoid arthritis (RA). Methods A systemic search was conducted from electronic databases (PubMed/Medline, Cochrane Library, and Google Scholar) from inception to 14th November 2023. All statistical analyses were conducted in Review Manager 5.4.1. Studies meeting inclusion criteria were selected. A random-effect model was used when heterogeneity was seen to pool the studies, and the result was reported in prevalence and their corresponding 95% confidence interval (CI). Other outcomes were assessed using qualitative analysis. Results A total of eight studies (six observational studies and 2 trials were used to conduct this systematic review and meta-analysis. A total population of 138,041 patients was used. Pooled analysis showed a statistically significant risk of GI bleeding in RA patients receiving NSAIDs (prevalence = 2% (1%, 3%); P < 0.00001; I2 = 98%). Qualitatively, causes and outcomes were discussed. Conclusion Our study showed that 2% RA patients were subjected to GI bleeding, when they used NSAIDs. Other causes of GI bleeding were age-related factors, cardiovascular events, history of GI complications, and peptic ulcers. Outcome varied by the use of specific NSAIDs and the presence of comorbidities. Recent guidelines for the management of RA may mention GI bleeding as a potential complication, but the level of emphasis placed on this issue varies. Some guidelines provide comprehensive recommendations for its prevention and management, while others offer limited guidance.
Collapse
Affiliation(s)
- Shobhit Piplani
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Vladimir Jelic
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Adejoke Johnson
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Usman Shah
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Shiny Kolli
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Steve Kong
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Nikola Tanasijevic
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Vishal Reddy Bejugam
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Sumaja Reddy Goguri
- Frank H. Netter MD School of Medicine/St. Vincent’s Medical Centre, Connecticut, United States
| | - Phanidhar Mogga
- Frank H. Netter MD School of Medicine/St. Vincent’s Medical Centre, Connecticut, United States
| | - Sripada Preetham Kasire
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | - Salil Chaturvedi
- Jacobi Medical Centre/North Central Bronx, Albert Einstein College of Medicine, NYC Health and Hospitals, New York, New York, United States
| | | |
Collapse
|
40
|
Gad HA, Abbas H, El Sayed NS, Khattab MA, El Hassab MA, Mansour M. Berberine loaded thermosensitive lipid nanoparticles: in vitro characterization, in silico study, and in vivo anti-arthritic effect. J Liposome Res 2024; 34:303-315. [PMID: 37856332 DOI: 10.1080/08982104.2023.2273390] [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: 05/30/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Thermoresponsive drug delivery systems have been used to treat diseases that cause hyperthermia or elevated body tissue temperatures, viz., rheumatoid arthritis and different cancers. The aim of the study was to enhance berberine (BER) release using thermosensitive nanostructured lipid carriers (TNLCs) through intra-articular administration for the management of arthritis. TNLCs were prepared using binary mixtures of stearic acid and decanoic acid as solid and liquid lipids, respectively. Lipid mixtures with an optimum melting point were assessed using differential scanning calorimetry studies. In vitro characterization of the BER TNLCs included particle size, zeta potential, entrapment efficiency, and drug release at 37 °C and 41 °C. Joint diameter measurement, real-time polymerase chain reaction (RT-PC) analysis, enzyme-linked immunosorbent assay (ELISA) for inflammatory markers, and histological evaluation of the dissected joints were all performed in vivo on rats with adjuvant-induced arthritis. In vitro characterization revealed negatively charged BER-loaded TNLCs with a spherical shape, particle size less than 500 nm, BER entrapment efficiency up to 79%, and a high drug release rate at an elevated temperature of 41 °C. In silico studies revealed the affinity of BER to different formula components and to the measured biomarkers. In vivo assessment of the optimum TNLCs showed that BER TNLCs were superior to the BER solution suspension regarding their effect on inflammatory biomarkers, joint diameter, and histological studies.
Collapse
Affiliation(s)
- Heba A Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Haidy Abbas
- Department of Pharmaceutics, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamed A Khattab
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Mahmoud A El Hassab
- Department of Medicinal Chemistry, Faculty of Pharmacy, King Salman International University (KSIU), Egypt
| | - Mai Mansour
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
41
|
Chen H, Fu X, Wu X, Zhao J, Qiu F, Wang Z, Wang Z, Chen X, Xie D, Huang J, Fan J, Yang X, Song Y, Li J, He D, Xiao G, Lu A, Liang C. Gut microbial metabolite targets HDAC3-FOXK1-interferon axis in fibroblast-like synoviocytes to ameliorate rheumatoid arthritis. Bone Res 2024; 12:31. [PMID: 38782893 PMCID: PMC11116389 DOI: 10.1038/s41413-024-00336-6] [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: 12/13/2023] [Revised: 03/18/2024] [Accepted: 04/07/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease. Early studies hold an opinion that gut microbiota is environmentally acquired and associated with RA susceptibility. However, accumulating evidence demonstrates that genetics also shape the gut microbiota. It is known that some strains of inbred laboratory mice are highly susceptible to collagen-induced arthritis (CIA), while the others are resistant to CIA. Here, we show that transplantation of fecal microbiota of CIA-resistant C57BL/6J mice to CIA-susceptible DBA/1J mice confer CIA resistance in DBA/1J mice. C57BL/6J mice and healthy human individuals have enriched B. fragilis than DBA/1J mice and RA patients. Transplantation of B. fragilis prevents CIA in DBA/1J mice. We identify that B. fragilis mainly produces propionate and C57BL/6J mice and healthy human individuals have higher level of propionate. Fibroblast-like synoviocytes (FLSs) in RA are activated to undergo tumor-like transformation. Propionate disrupts HDAC3-FOXK1 interaction to increase acetylation of FOXK1, resulting in reduced FOXK1 stability, blocked interferon signaling and deactivation of RA-FLSs. We treat CIA mice with propionate and show that propionate attenuates CIA. Moreover, a combination of propionate with anti-TNF etanercept synergistically relieves CIA. These results suggest that B. fragilis or propionate could be an alternative or complementary approach to the current therapies.
Collapse
Affiliation(s)
- Hongzhen Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Xuekun Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Xiaohao Wu
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Division of Immunology and Rheumatology, Stanford University, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Junyi Zhao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Fang Qiu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Zhenghong Wang
- Institute of Plant and Food Science, Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhuqian Wang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Xinxin Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
| | - Duoli Xie
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Jie Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China
| | - Junyu Fan
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yi Song
- Institute of Plant and Food Science, Department of Biology, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jie Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dongyi He
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, 510006, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055, China.
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, 999077, China.
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China.
| |
Collapse
|
42
|
Inchingolo F, Inchingolo AM, Fatone MC, Avantario P, Del Vecchio G, Pezzolla C, Mancini A, Galante F, Palermo A, Inchingolo AD, Dipalma G. Management of Rheumatoid Arthritis in Primary Care: A Scoping Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:662. [PMID: 38928909 PMCID: PMC11203333 DOI: 10.3390/ijerph21060662] [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: 02/29/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024]
Abstract
Rheumatoid arthritis (RA) can lead to severe joint impairment and chronic disability. Primary care (PC), provided by general practitioners (GPs), is the first level of contact for the population with the healthcare system. The aim of this scoping review was to analyze the approach to RA in the PC setting. PubMed, Scopus, and Web of Science were searched using the MESH terms "rheumatoid arthritis" and "primary care" from 2013 to 2023. The search strategy followed the PRISMA-ScR guidelines. The 61 articles selected were analyzed qualitatively in a table and discussed in two sections, namely criticisms and strategies for the management of RA in PC. The main critical issues in the management of RA in PC are the following: difficulty and delay in diagnosis, in accessing rheumatological care, and in using DMARDs by GPs; ineffective communication between GPs and specialists; poor patient education; lack of cardiovascular prevention; and increase in healthcare costs. To overcome these criticisms, several management strategies have been identified, namely early diagnosis of RA, quick access to rheumatology care, effective communication between GPs and specialists, active patient involvement, screening for risk factors and comorbidities, clinical audit, interdisciplinary patient management, digital health, and cost analysis. PC appears to be the ideal healthcare setting to reduce the morbidity and mortality of chronic disease, including RA, if a widespread change in GPs' approach to the disease and patients is mandatory.
Collapse
Affiliation(s)
- Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | | | - Pasquale Avantario
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | - Gaetano Del Vecchio
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | - Carmela Pezzolla
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | | | - Andrea Palermo
- College of Medicine and Dentistry, Birmingham B4 6BN, UK
| | - Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (P.A.); (G.D.V.); (C.P.); (A.M.); (A.D.I.); (G.D.)
| |
Collapse
|
43
|
Wang Y, Guo R, Zou M, Jiang L, Kong L, Zhao S, Zhang X, Wang W, Xu B. Combined ROS Sensitive Folate Receptor Targeted Micellar Formulations of Curcumin Effective Against Rheumatoid Arthritis in Rat Model. Int J Nanomedicine 2024; 19:4217-4234. [PMID: 38766660 PMCID: PMC11100960 DOI: 10.2147/ijn.s458957] [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: 01/10/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
Introduction Rheumatoid arthritis (RA) is an inflammatory immune-mediated disease that involves synovitis, cartilage destruction, and even joint damage. Traditional agents used for RA therapy remain unsatisfactory because of their low efficiency and obvious adverse effects. Therefore, we here established RA microenvironment-responsive targeted micelles that can respond to the increase in reactive oxygen species (ROS) levels in the joint and improve macrophage-specific targeting of loaded drugs. Methods We here prepared ROS-responsive folate-modified curcumin micelles (TK-FA-Cur-Ms) in which thioketal (TK) was used as a ROS-responsive linker for modifying polyethylene glycol 5000 (PEG5000) on the micellar surface. When micelles were in the ROS-overexpressing inflammatory microenvironment, the PEG5000 hydration layer was shed, and the targeting ligand FA was exposed, thereby enhancing cellular uptake by macrophages through active targeting. The targeting, ROS sensitivity and anti-inflammatory properties of the micelles were assessed in vitro. Collagen-induced arthritis (CIA) rats model was utilized to investigate the targeting, expression of serum inflammatory factors and histology change of the articular cartilage by micelles in vivo. Results TK-FA-Cur-Ms had a particle size of 90.07 ± 3.44 nm, which decreased to 78.87 ± 2.41 nm after incubation with H2O2. The micelles exhibited in vitro targeting of RAW264.7 cells and significantly inhibited inflammatory cytokine levels. Pharmacodynamic studies have revealed that TK-FA-Cur-Ms prolonged the drug circulation and exhibited augmented cartilage-protective and anti-inflammatory effects in vivo. Conclusion The unique ROS-responsive targeted micelles with targeting, ROS sensitivity and anti-inflammatory properties were successfully prepared and may offer an effective therapeutic strategy against RA.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
| | - Ming Zou
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Lingling Jiang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, People’s Republic of China
| | - Sen Zhao
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Xuan Zhang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Wei Wang
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| | - Baoli Xu
- Department of Pharmacy, Affiliated Zhongshan Hospital of Dalian University, Dalian, People’s Republic of China
| |
Collapse
|
44
|
Yang X, Zhao L, Pang Y. cGAS-STING pathway in pathogenesis and treatment of osteoarthritis and rheumatoid arthritis. Front Immunol 2024; 15:1384372. [PMID: 38765007 PMCID: PMC11099256 DOI: 10.3389/fimmu.2024.1384372] [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: 02/09/2024] [Accepted: 04/12/2024] [Indexed: 05/21/2024] Open
Abstract
Osteoarthritis (OA) and Rheumatoid Arthritis (RA) are significant health concerns with notable prevalence and economic impact. RA, affecting 0.5% to 1.0% of the global population, leads to chronic joint damage and comorbidities. OA, primarily afflicting the elderly, results in joint degradation and severe pain. Both conditions incur substantial healthcare expenses and productivity losses. The cGAS-STING pathway, consisting of cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING), is a crucial component of mammalian immunity. This pathway is responsible for detecting foreign DNA, particularly double-stranded DNA (dsDNA), triggering innate immune defense responses. When cGAS recognizes dsDNA, it catalyzes the synthesis of cyclic GMP-AMP (cGAMP), which then binds to and activates STING. Activated STING, in turn, initiates downstream signaling events leading to the production of interferons and other immune mediators. The cGAS-STING pathway is essential for defending against viral infections and maintaining cellular balance. Dysregulation of this pathway has been implicated in various inflammatory diseases, including arthritis, making it a target for potential therapeutic interventions. Understanding the intricate molecular signaling network of cGAS-STING in these arthritis forms offers potential avenues for targeted therapies. Addressing these challenges through improved early detection, comprehensive management, and interventions targeting the cGAS-STING pathway is crucial for alleviating the impact of OA and RA on individuals and healthcare systems. This review offers an up-to-date comprehension of the cGAS-STING pathway's role in the development and therapeutic approaches for these arthritis types.
Collapse
Affiliation(s)
- XiCheng Yang
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, China
| | - LiLi Zhao
- Orthopedics and Arthrology, People Hospital of Xingtai, Xingtai, Hebei, China
| | - YinQuan Pang
- Graduate School, Chengde Medical University, Chengde, Hebei, China
| |
Collapse
|
45
|
Cheng Y, Ji Y, Feng K, Zhang X, Xiao Y, Jing W. A study of the molecular mechanism of action of Jiawei Guizhishaoyaozhimu Decoction during rheumatoid arthritis therapy based on basic of network pharmacology and experimental verification. Exp Ther Med 2024; 27:212. [PMID: 38590567 PMCID: PMC11000449 DOI: 10.3892/etm.2024.12499] [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: 06/10/2023] [Accepted: 02/16/2024] [Indexed: 04/10/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease, which primarily affects the joints. The aim of the present study was to predict the main active ingredients of Jiawei Guizhishaoyaozhimu Decoction (JWGZSYZMD) and potential targets of this treatment during RA therapy by using molecular docking and network pharmacology methods. In addition, another aim was to investigate the therapeutic effects and mechanism of JWGZSYZMD on joint inflammation in rat models of collagen Ⅱ-induced arthritis (CIA). JWGZSYZMD ingredients and targets and genes associated with RA first extracted from traditional Chinese medicine (TCM) Systems Pharmacology Database and Analysis Platform, Bioinformatics Analysis Tool of Molecular Mechanism-TCM and Genecards databases, which were then transferred to the STRING database to set up protein interaction networks. The crystal structures of target proteins were also downloaded from the Protein Data Bank before molecular docking of compounds onto the protein targets was performed using AutoDock Vina software. In addition, a drug compound target visualization network was constructed using Cytoscape 3.7.2 software, which was used to elucidate the main mechanism underlying the anti-RA effect of JWGZSYZMD. A CIA rat model was established and animals were divided into the control, CIA model, JWGZSYZMD treatment (low-, medium- and high-dose) and tripterygium glycoside groups. Compared with the rats in the CIA model group, the joint scores of the rats in the high-dose group of JWGZSYZMD were significantly lower after 21 days of treatment. The expression levels of IL-6, TNF-α, IL-1β and IL-17A in the synovial supernatant of the model rats were lower compared with those in the CIA group. Also, the expression of the aforementioned cytokines in the high-dose JWGZSYZMD group was significantly lower compared with those in the CIA model group. To conclude, using molecular docking combined with network pharmacology, the material basis and molecular mechanism underlying the effects of JWGZSYZMD during RA therapy were studied, which could potentially provide a reference for future clinical applications.
Collapse
Affiliation(s)
- Yiji Cheng
- Department of Rheumatology and Immunology, Beijing University Of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| | - Yue Ji
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Kaidi Feng
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Xinyuan Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| | - Yunming Xiao
- State Key Laboratory of Kidney Diseases, First Medical Center of Chinese People's Liberation Army General Hospital, Medical School of Chinese People's Liberation Army, Beijing 100000, P.R. China
| | - Weixia Jing
- Department of Rheumatology and Immunology, Beijing University Of Chinese Medicine Third Affiliated Hospital, Beijing 100029, P.R. China
| |
Collapse
|
46
|
Dai Y, Chen L, Zhang Z, Liu X. Identification and validation of immune-related genes in osteoarthritic synovial fibroblasts. Heliyon 2024; 10:e28330. [PMID: 38571590 PMCID: PMC10988018 DOI: 10.1016/j.heliyon.2024.e28330] [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: 07/10/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
Objective OA was generally considered as a non-inflammatory disease dominated by articular cartilage degeneration. However, the role of synovitis in OA pathogenesis has received increasing attention. Recent studies support that OA patients have a pro-inflammatory/catabolic synovial environment similar to RA patients, promoting the occurrence and development of OA. Therefore, we investigated the co-immune-related genes and pathways of OA and RA to explore whether part of the pathogenesis of RA synovitis can be used to explain OA synovitis. Methods Data of GSE29746 and GSE12021 were downloaded from the Gene Expression Omnibus (GEO) database. Compared with control group, differentially expressed genes (DEGs) of OA and RA groups were screened separately by R software, Venny website was used to screen co-DEGs. Metascape was used to screen the common enriched terms and pathways between OA and RA. STRING website and Cytoscape software were used to map protein-protein interaction (PPI) networks and screen co-hub genes. GSE29746 was selected as the test dataset, and GSE12021 as the validation dataset for validate the co-hub genes. The results were validated by western blotting (WB) and real-time quantitative polymerase chain reaction (qPCR) of clinical synovial samples. Results We identified 573 OA-related DEGs, 148 RA-related DEGs, and 52 co-DEGs, revealing 14 common enriched terms, most of which were related to immune inflammation. IL7R was the only upregulated co-hub gene between OA and RA in the PPI network, consistent with the validation dataset. IL7R was highly expressed in clinical osteoarthritic synovial samples (P < 0.001). Conclusion Our findings suggested that IL7R is a critical co-DEG in OA and RA and confirmed the involvement of immune inflammation in disease pathogenesis. Furthermore, it confirms the role of IL7R in synovial inflammation in RA and OA synovitis and provides evidence for further investigation of OA immune inflammation.
Collapse
Affiliation(s)
- Yaduan Dai
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Chen
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhan Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xueyong Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
47
|
Liu XR, Li SF, Mei WY, Liu XD, Zhou RB. Isorhamnetin Downregulates MMP2 and MMP9 to Inhibit Development of Rheumatoid Arthritis through SRC/ERK/CREB Pathway. Chin J Integr Med 2024; 30:299-310. [PMID: 38212502 DOI: 10.1007/s11655-023-3753-6] [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] [Accepted: 06/29/2023] [Indexed: 01/13/2024]
Abstract
OBJECTIVE To investigate the effect of isorhamnetin on the pathology of rheumatoid arthritis (RA). METHODS Tumor necrosis factor (TNF)- α -induced fibroblast-like synoviocytes (FLS) was exposed to additional isorhamnetin (10, 20 and 40 µ mol/L). Overexpression vectors for matrix metalloproteinase-2 (MMP2) or MMP9 or SRC were transfected to explore their roles in isorhamnetin-mediated RA-FLS function. RA-FLS viability, migration, and invasion were evaluated. Moreover, a collagen-induced arthritis (CIA) rat model was established. Rats were randomly divided to sham, CIA, low-, medium-, and high-dosage groups using a random number table (n=5 in each group) and administed with normal saline or additional isorhamnetin [2, 10, and 20 mg/(kg·day)] for 4 weeks, respectively. Arthritis index was calculated and synovial tissue inflammation was determined in CIA rats. The levels of MMP2, MMP9, TNF-α, interleukin-6 (IL-6), and IL-1 β, as well as the phosphorylation levels of SRC, extracellular regulated kinase (ERK), and cyclic adenosine monophosphate response element-binding (CREB), were detected in RA-FLS and synovial tissue. Molecular docking was also used to analyze the binding of isorhamnetin to SRC. RESULTS In in vitro studies, isorhamnetin inhibited RA-FLS viability, migration and invasion (P<0.05). Isorhamnetin downregulated the levels of MMP2, MMP9, TNF-α, IL-6, and IL-1 β in RA-FLS (P<0.05). The overexpression of either MMP2 or MMP9 reversed isorhamnetin-inhibited RA-FLS migration and invasion, as well as the levels of TNF-α, IL-6, and IL-1 β (P<0.05). Furthermore, isorhamnetin bound to SRC and reduced the phosphorylation of SRC, ERK, and CREB (P<0.05). SRC overexpression reversed the inhibitory effect of isorhamnetin on RA-FLS viability, migration and invasion, as well as the negative regulation of MMP2 and MMP9 (P<0.05). In in vivo studies, isorhamnetin decreased arthritis index scores (P<0.05) and alleviated synovial inflammation. Isorhamnetin reduced the levels of MMP2, MMP9, TNF-α, IL-6, and IL-1 β, as well as the phosphorylation of SRC, ERK, and CREB in synovial tissue (P<0.05). Notably, the inhibitory effect of isorhamnetin was more pronounced at higher concentrations (P<0.05). CONCLUSION Isorhamnetin exhibited anti-RA effects through modulating SRC/ERK/CREB and MMP2/MMP9 signaling pathways, suggesting that isorhamnetin may be a potential therapeutic agent for RA.
Collapse
Affiliation(s)
- Xiao-Rong Liu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Shuo-Fu Li
- Department of Orthopaedics, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Wen-Ya Mei
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiang-Dan Liu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ri-Bao Zhou
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| |
Collapse
|
48
|
Wang G, Xu YL, Zhang XH, Tang L, Li Y. LncRNA HOTTIP regulates TLR4 promoter methylation by recruiting H3K4 methyltransferase MLL1 to affect apoptosis and inflammatory response of fibroblast-like synoviocyte in rheumatoid arthritis. Kaohsiung J Med Sci 2024; 40:335-347. [PMID: 38363110 DOI: 10.1002/kjm2.12805] [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: 08/29/2023] [Revised: 12/05/2023] [Accepted: 12/24/2023] [Indexed: 02/17/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease, and the role of HOXA transcript at the distal tip (HOTTIP) in its pathogenesis remains underexplored. This study investigates the mechanism by which HOTTIP influences apoptosis and the inflammatory response of fibroblast-like synoviocytes (FLS). An RA mouse model was established, and clinical scores were analyzed. Pathological changes in synovial tissues, bone mineral density (BMD) of the paws, serum tartrate-resistant acid phosphatase (TRAP) activity, and TNF-α and IL-1β levels were assessed. FLS were transfected, and cell proliferation and apoptosis were examined. The RNA-pull-down assay determined HOTTIP's interaction with mixed-lineage leukemia 1 (MLL1), while RNA immunoprecipitation assay measured HOTTIP expression pulled down by MLL1. The levels of MLL1 and toll-like receptor 4 (TLR4) after MLL1 overexpression based on HOTTIP silencing were determined. Chromatin immunoprecipitation (ChIP) was performed with H3K4me3 as an antibody, followed by the evaluation of TLR4 expression. HOTTIP expression was elevated in RA mouse synovial tissues. Inhibition of HOTTIP led to reduced clinical scores, inflammatory infiltration, synovial hyperplasia, TRAP activity, and TNF-α and IL-1β levels, along with increased BMD. In vitro Interference with HOTTIP suppressed RA-FLS apoptosis and inflammation. HOTTIP upregulated TLR4 expression by recruiting MLL1 to facilitate TLR4 promoter methylation. MLL1 overexpression reversed HOTTIP silencing-mediated repression of RA-FLS apoptosis. Activation of H3K4 methylation counteracted HOTTIP knockout, ameliorating the inflammatory response. HOTTIP regulates TLR4 expression by recruiting MLL1, leading to TLR4 promoter methylation, thereby suppressing RA-FLS proliferation and inducing cell apoptosis and inflammatory response in RA.
Collapse
Affiliation(s)
- Guan Wang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Yu-Lin Xu
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Xi-Hai Zhang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Lian Tang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Sichuan Provincial Laboratory of Orthopaedic Engineering Luzhou, Sichuan, China
| | - Yao Li
- Laboratory Animal Center, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
49
|
Carrascosa JM, Echarri A, Gavín Sebastián O, García de la Peña P, Martínez Pérez O, Ramirez S, Valderrama M, Montoro Álvarez M. Contraceptive Recommendations for Women with Immune-Mediated Inflammatory Diseases: A Delphi Consensus. Adv Ther 2024; 41:1372-1384. [PMID: 38326688 DOI: 10.1007/s12325-023-02779-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/21/2023] [Indexed: 02/09/2024]
Abstract
INTRODUCTION Immune-mediated inflammatory diseases (IMID) are a group of disorders characterized by chronic inflammation caused by an altered immune regulation in targeted organs or systems. IMID itself could have an implied increased risk of venous thromboembolism (VTE) and this risk varies throughout the course of the disease as well as with some contraceptive methods and treatments. The aim of this study was to present some key considerations in relation to contraception in women with IMID. METHODS This was an exploratory study conducted in Spain following the online modified Delphi methodology with two rounds of participation. Four questionnaires were designed for each medical specialty: gastroenterology, rheumatology, dermatology, and gynecology. Each questionnaire was divided in three domains: general recommendations about IMID, specific recommendations, and contraceptive methods for patients with IMID. A 5-point Likert scale measured agreement with each statement, with an 80% agreement threshold. Following the first round, the percentage of each response was calculated for every item. Subsequently, a second round was conducted to reach a consensus on the items for which discrepancies were observed. RESULTS A total of 52 and 50 experts participated in the first and second round, respectively. Participants agreed on the existence of a higher risk of VTE in inflammatory bowel diseases, psoriasis, and rheumatoid arthritis diseases. Regarding recommendations for contraceptive methods in patients with IMID, experts considered the hormonal intrauterine device (IUD) as a first-line contraceptive (80.0%) and low doses of progesterone-only pills if the latter is not recommended (88.0%). Most of the interviewees concurred on the importance of the patients' contraceptive needs during the disease course (98.1%). CONCLUSION Raising awareness and promoting a multidisciplinary relationship among the physicians involved in the therapeutic decisions by considering all the risk factors when prescribing a contraceptive method is important to prevent VTE in women with IMID.
Collapse
Affiliation(s)
- José Manuel Carrascosa
- Servicio de Dermatología, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, IGTP, Badalona, Spain
| | - Ana Echarri
- Servicio de Digestivo, Complejo Hospitalario Universitario de Ferrol, La Coruña, Spain
| | - Olga Gavín Sebastián
- Servicio de Hematología y Hemoterapia, Hospital Clínico Universitario Lozano Blesa, Saragossa, Spain
| | | | - Oscar Martínez Pérez
- Servicio de Obstetricia y Ginecología, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Susan Ramirez
- Pfizer Medical SLU, Av. de Europa, 20, B, Alcobendas, 28108, Madrid, Spain
| | - Mónica Valderrama
- Pfizer Medical SLU, Av. de Europa, 20, B, Alcobendas, 28108, Madrid, Spain
| | | |
Collapse
|
50
|
Ahmed EA, Alzahrani AM, Abdelsalam SA, Ibrahim HIM. Flavipin from fungi as a potential inhibitor of rheumatoid arthritis signaling molecules. Inflammopharmacology 2024; 32:1171-1186. [PMID: 38349589 DOI: 10.1007/s10787-024-01429-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/02/2024] [Indexed: 04/11/2024]
Abstract
Flavipin, a fungal lower molecular weight biomolecule (MW 196.16 g/mol), has not been yet extensively studied for beneficial preclinical and clinical applications. In recent years, various preclinical mouse models including adjuvant-induced arthritis (AIA) were employed to understand mechanisms associated with Rheumatoid arthritis (RA) and to develop new therapeutic drugs. In the current study, we studied the inhibitory effect of Flavipin on major signaling molecules involved in the inflammatory response during RA using both in-silico virtual interaction and in vivo mouse model of AIA. Our in-silico results clarified that Flavipin interacts with the tumor necrosis factor alpha (TNF-α) through conventional hydrogen binding (H-H) at one of TNF-α critical amino acids tyrosine residues, Tyr119, with binding energy (b.e.) -5.9. In addition, Flavipin binds to ATP-binging sites of the Jesus kinases, JAK1, JAK2 and JAK3, through H-H (b. e. between -5.8 and -6.1) and then it may inhibit JAKs, regulators of RA signaling molecules. Moreover, our molecular dynamics stimulation for the docked TNF-α/Flavipin complex confirmed the specificity and the stability of the interaction. In vitro, Flavipin is not toxic to normal cells at doses below 50 µM (its IC50 in normal fibroblast cell line was above 100 µM). However, in vivo, the arthritis score and hind paw oedema parameters were modulated in Flavipin treated mice. Consistent with the in-silico results the levels of the TNF-α, the nuclear transcription factor kappaB (NF-κB) and the signal transduction and activator of transcription (STAT3, downstream of JAKs) were modulated at joint tissues of the hind-paw of Flavipin/AIA treated mice. Our data suggest Flavipin as a potential therapeutic agent for arthritis can inhibit RA major signaling molecules.
Collapse
Affiliation(s)
- Emad A Ahmed
- Department of Biological Sciences, College of Science, King Faisal University, 31982, Hofouf, Alhasa, Saudi Arabia.
- Laboratory of Molecular Physiology, Zoology Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt.
| | - Abdulaah M Alzahrani
- Department of Biological Sciences, College of Science, King Faisal University, 31982, Hofouf, Alhasa, Saudi Arabia
| | - Salah A Abdelsalam
- Department of Biological Sciences, College of Science, King Faisal University, 31982, Hofouf, Alhasa, Saudi Arabia
- Department of Zoology, Faculty of Science, Assiut University, Asyut, 71516, Egypt
| | - Hairul-Islam M Ibrahim
- Department of Biological Sciences, College of Science, King Faisal University, 31982, Hofouf, Alhasa, Saudi Arabia
- Pondicherry Centre for Biological Science and Educational Trust, Kottakuppam, Pondicherry, 605104, India
| |
Collapse
|