1
|
Ma Z, Wu S, Xiong LE, Zhong J, Lin B, Chen L, Xiong T, Wu Y. Interaction of nutritional and inflammatory levels on all-cause mortality among individuals with rheumatoid arthritis: a prospective cohort study. Clin Rheumatol 2025:10.1007/s10067-025-07458-0. [PMID: 40295462 DOI: 10.1007/s10067-025-07458-0] [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: 10/22/2024] [Revised: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Malnutrition and systemic inflammation are frequently prevalent in individuals with rheumatoid arthritis (RA). However, limited studies have explored the combined impacts of nutritional and inflammatory levels on all-cause mortality among RA. This study is aimed at investigating these potential associations. METHODS We involved 2213 RA patients from the National Health and Nutrition Examination Survey (2001-2018). Nutritional risk index (NRI) and systemic inflammatory response index (SIRI) were used to evaluate the nutritional and inflammatory status of participants, respectively. Weight-based Kaplan-Meier survival curves and COX proportional hazard models were employed to ascertain the independent and joint association. RESULTS Over a median follow-up of 7.98 years, 544 deaths occurred. Following the adjustment for confounding factors, we found that individuals with moderate/severe malnutrition (HR, 2.13; 95%CI, 1.50-3.03) or high SIRI (HR, 1.49; 95%CI, 1.07-2.07) were independently associated with an increased risk of all-cause mortality. Furthermore, the simultaneous moderate/severe malnutrition and high SIRI further elevated the mortality risk (HR, 2.34; 95%CI, 1.53-3.56). An additive interaction was observed between malnutrition and high SIRI, yielding an excess risk of 0.34 (95%CI 0.06-0.62, P = 0.014), and the attributable proportion for the interaction was 20.9% (95%CI 7.6-34.2%). CONCLUSIONS This research indicated that malnutrition and high systemic inflammation levels were independent risk factors for the prognosis of RA patients, and co-occurrence can further deteriorate the prognosis. Our findings highlight the importance of maintaining nutrition and anti-inflammation in patients with RA, providing new insights into the prognosis of RA patients.
Collapse
Affiliation(s)
- Zhuang Ma
- School of Public Health, Guangzhou Medical University, Guangzhou, 51136, China
| | - Shixin Wu
- School of Public Health, Guangzhou Medical University, Guangzhou, 51136, China
| | - Lin-En Xiong
- School of Public Health, Guangzhou Medical University, Guangzhou, 51136, China
| | - Juncheng Zhong
- School of Public Health, Guangzhou Medical University, Guangzhou, 51136, China
| | - Baiwen Lin
- School of Public Health, Guangzhou Medical University, Guangzhou, 51136, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety and the Ministry of Education (MOE) Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Ting Xiong
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuanjue Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
2
|
Wasim R, Sumaiya, Ahmad A, Anwar A, Salman A. Microbial imbalance in the gut: a new frontier in Rheumatoid arthritis research. Inflammopharmacology 2025:10.1007/s10787-025-01737-7. [PMID: 40220199 DOI: 10.1007/s10787-025-01737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
A chronic autoimmune illness that causes joint destruction and inflammation, rheumatoid arthritis (RA) often results in disability. Genetic, environmental, and immune system variables all have a role in the pathophysiology of RA. The complex community of bacteria that live in the gastrointestinal system, known as the gut microbiota, has been implicated in the onset and progression of RA in recent years, according to mounting data. An imbalance in the gut microbiota's composition, known as dysbiosis, has been noted in RA patients. This imbalance may impact inflammatory pathways and immunological responses, which in turn may contribute to the development and severity of the illness. Research has shown that some bacterial species, including Firmicutes, Bacteroidetes, and Proteobacteria, are either more abundant or less prevalent in RA patients than in healthy people. The gut-immune system axis may be modulated, immunological tolerance may be affected, and pro-inflammatory cytokine production may be enhanced by these microbial changes, all of which may lead to systemic inflammation linked to RA. Moreover, changes in intestinal permeability and a rise in microbial metabolite translocation may make autoimmune reactions worse. Probiotics, antibiotics, and dietary changes have also been investigated as possible treatment approaches to help RA patients regain the balance of their gut microbiota. Still up for debate, however, are the precise ways in which the gut microbiome affects RA. Comprehending the complex connection between gut microbiota and RA may give new perspectives on managing and preventing the condition, as well as future prospects for medicines that target the microbiome.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India.
| | - Sumaiya
- Career Post Graduate Institute of Dental Sciences and Hospital, Lucknow, India
| | - Asad Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| | - Aamir Anwar
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| | - Aimen Salman
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| |
Collapse
|
3
|
Li M, Sun Y, Shan X, Tong Y, Fu Y, Ma X, Sun Z, Xiang Y, Zhu Y, Wang T, Wang X, Zhang J, Niu D. Roles of Immunity and Endogenous Retroelements in the Pathogenesis of Rheumatoid Arthritis and Treatment Strategies. Funct Integr Genomics 2025; 25:85. [PMID: 40205241 DOI: 10.1007/s10142-025-01583-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic, inflammatory, autoimmune disease that primarily affects the joints. RA usually results in synovial hyperplasia, expansion of "pannus" and destruction of cartilage. The etiology and pathogenesis of RA are not fully understood, but immunity has been shown to play an important role in the development of autoimmune diseases such as RA. In addition, endogenous retroelements, the remnants of ancient retroviruses in the human genome, are involved in cancer and/or immune disorders. As evidenced by increasing evidences that the aberrant expression of retroelements induces innate immunity, despite the fact that the expression of most retroelements has been epigenetically suppressed over a long period of evolution. With the continuous development of disease-modifying anti-rheumatic drugs (DMARDs), RA disease activity has been alleviated and improved. Unfortunately, some patients have a limited response to DMARDs, and the drugs also have the disadvantages of some side effects and high economic costs. This review summarizes the pathogenic mechanisms of RA and endogenous retroelements in autoimmunity, and concludes with a summary of treatments for RA, along with new therapeutic recommendations.
Collapse
Affiliation(s)
- Mingyang Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Yuanyuan Sun
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Xueting Shan
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Yuhong Tong
- Fourth School of Clinical Medicine, Zhejiang University of Traditional Chinese Medicine, Hangzhou, 310053, China
| | - Yite Fu
- Nanjing Outstanding Gene Technology Co, Nanjing, 210018, Jiangsu, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China
| | - Zhongxin Sun
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China
| | - Yun Xiang
- Jinhua Academy of Agricultural Sciences, Jinhua, 321000, Zhejiang, China
| | - Yidan Zhu
- Jinhua Academy of Agricultural Sciences, Jinhua, 321000, Zhejiang, China
| | - Tao Wang
- Nanjing Outstanding Gene Technology Co, Nanjing, 210018, Jiangsu, China
| | - Xin Wang
- Department of Plastic, Reconstructive & Hand Microsurgery, Ningbo NO.6 Hospital, Ningbo, 315000, Zhejiang, China.
| | - Jufang Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, 666 Wusu Street, Hangzhou, 311300, Zhejiang, China.
| |
Collapse
|
4
|
Ziyadullaev SK, Khudaiberdiev SS, Aripova TU, Chirumbolo S, Kamalov ZS, Bjørklund G, Rizaev JA, Tashkenbaeva EN, Khamidov OA, Gaffarov UB. Synovial Fluid as a Crucial Component of the Joint Microenvironment in Rheumatoid Arthritis. Immune Netw 2025; 25:e2. [PMID: 40342839 PMCID: PMC12056296 DOI: 10.4110/in.2025.25.e2] [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: 05/23/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 05/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease closely associated with synovial tissue proliferation, pannus formation in small joints such as the hands, wrists, and feet, cartilage destruction, and systemic complications such as pulmonary, cardiovascular, neurological, and skeletal muscle lesions, glucocorticoid-induced osteoporosis and infections. The importance of confirming the diagnosis and determining local activity is given to the study of synovial fluid. A deep understanding of the pathological process in the joint in RA, characterized by changes in autoreactive CD4+ T cells, B cells, macrophages, inflammatory cytokines, chemokines, and autoantibodies, has now been achieved, although much remains to be explored. This article provides an updated overview of the pathogenesis of RA, revealing even more therapeutic targets for the intra-articular pathological process.
Collapse
Affiliation(s)
| | | | | | | | | | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana 8622, Norway
| | | | | | - Obid Abdurakhmanovich Khamidov
- Rehabilitology and Sports Medicine Research Institute of the Samarkand State Medical University, Samarkand 140100, Uzbekistan
| | | |
Collapse
|
5
|
Liu C, Zhao S, Qiao L, Ren Y, Liu K, Bi S, Li B, Yuan A, Zheng L, Wang Z, Xu Z, Zhang Y. A druggable targets discovery strategy for diseases (DTDS): Taking Rheumatoid arthritis as a case. Int Immunopharmacol 2025; 149:114182. [PMID: 39904046 DOI: 10.1016/j.intimp.2025.114182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 01/24/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025]
Abstract
Identifying effective druggable targets with disease-specific for diseases is a tremendous challenge in new drug development. However, current studies of druggable targets identification are most based on either druggability or disease-specific, lacking a combination of two factors. To further improve the accuracy of druggable targets discovery, a druggable target discovery strategy for diseases (DTDS) was proposed, which combined druggable targets prediction by machine learning and key targets identification by tissue-level and cellular-level transcriptomics analysis. Rheumatoid arthritis (RA), an autoimmune disease that cannot be treated entirely, was taken as a case. First, the protein-protein interaction network was constructed as the disease background network, and the classification models were established based on the topological parameters of known RA-druggable targets with druggability and non-RA targets without therapeutic effects on RA. 168 potential druggable targets were predicted by the classification models from 264 RA-related targets. Subsequently, 40 RA-specific targets were identified by tissue-level and cellular-level transcriptomics analysis from 168 potential druggable targets. Most of them were RA-druggable targets except PSMB9 and PTPRC. Finally, PSMB9 and PTPRC were further verified by in vitro experiments. The results showed that the inhibitor of PSMB9 or PTPRC could effectively inhibit inflammation and abnormal proliferation of synovial cells, proving that PSMB9 and PTPRC were potential RA-druggable targets, and further indicating that DTDS had high accuracy. In conclusion, the DTDS strategy established in this study is reliable and has been proven in identification of potential RA-druggable targets, which is expected to provide ideas and methods for systematic discovery of potential druggable targets for diseases.
Collapse
Affiliation(s)
- Chaoqun Liu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shuai Zhao
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Liansheng Qiao
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yue Ren
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Kaiyang Liu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shijie Bi
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Beiyan Li
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Anlei Yuan
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lulu Zheng
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zewen Wang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhenzhen Xu
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yanling Zhang
- Key Laboratory of TCM-information Engineer of State Administration of TCM, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
6
|
Buttari B, Recalchi S, Riitano G, Capozzi A, Ucci FM, Manganelli V, Fratini F, Profumo E, Garofalo T, Alessandri C, Misasi R, Conti F, Longo A, Sorice M. Extracellular microvesicles from patients with Rheumatoid arthritis promote dendritic cell activation in vitro. Front Immunol 2025; 16:1532114. [PMID: 40109338 PMCID: PMC11920144 DOI: 10.3389/fimmu.2025.1532114] [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: 11/21/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Rheumatoid Arthritis (RA) is a systemic autoimmune disease characterized by chronic synovial inflammation affecting diarthrodial joints, with cartilage destruction and bone erosion. Environmental inflammatory stimuli can induce maturation of dendritic cells (DCs), which promote differentiation and activation of effector T lymphocytes. We previously highlighted the role of extracellular microvesicles (EMVs) in pathogenesis by carrying antigens that trigger autoantibody production. In this investigation we verified whether EMVs may activate immature monocyte-derived DCs, inducing phenotypic and functional characteristics of mature DCs. Methods EMVs were obtained from 7 RA patients naïve to biological disease-modifying anti-rheumatic drugs (DMARDs) and tested for their capability to activate DCs from healthy donors. Results We preliminary confirmed by western blot that carbamylated and citrullinated proteins are present in EMVs from RA patients. Moreover, surface marker phenotyping indicated that EMV treated-DCs exhibit increased expression of CD83 and CD86, as well as of CD83+ HLA-DR+ CD80+ CD86+ cells, indicating that the DCs are in a mature state. Furthermore, biochemical data demonstrated that EMVs from plasma of RA patients induce MAPK and NF-κB activation in DCs. EMVs from the plasma of RA patients were also able to stimulate DCs to produce IL-12, IL-1β and IL-10, inducing a proinflammatory phenotype. Conclusions These findings demonstrate that EMVs from RA patients promote DC activation in vitro, suggesting a potential mechanism by which RA microenvironment perpetuates inflammation through the modulation of DC function. These knowledges provide new insight in the role of EMVs in the pathogenesis of RA and their potential role as therapeutic targets.
Collapse
Affiliation(s)
- Brigitta Buttari
- Department of Cardiovascular and Endocrine-metabolic Diseases, and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Serena Recalchi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Antonella Capozzi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Federica Maria Ucci
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Federica Fratini
- Proteomics Core Facility, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-metabolic Diseases, and Aging, Istituto Superiore di Sanità, Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Cristiano Alessandri
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Fabrizio Conti
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Agostina Longo
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
7
|
Rafati A, Ramezani R, Esmaeili Gouvarchin Ghaleh H, Bahrami S, Alvanegh AG, Masoudi MR. Calcitriol Treated Mesenchymal Stem Cells Modulated Immune Response in Collagen-Induced Rheumatoid Arthritis in BALB/c Mice. Transplant Proc 2025; 57:355-363. [PMID: 39837674 DOI: 10.1016/j.transproceed.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND AND AIM Rheumatoid arthritis (RA) is a chronic inflammatory disease that primarily involves synovial joints. During the past decade, disease-modifying antirheumatic drugs and biologic agents have been introduced for the treatment of RA. However, they have limitations, including incomplete treatment response, adverse effects requiring drug withdrawal, fall off in efficacy over time, high cost of biologic agents, and refractory cases. Consequently, there is a need to establish safe and effective advanced therapeutic modalities for RA to overcome the shortcomings of current treatments. METHODS MSCs after isolation were exposed to 200 nM calcitriol. Rheumatoid arthritis was induced in BALB/c mice using collagen and Freund's complete adjuvant. One week after immunization, the mice were divided into 3 groups including without treatment, groups treated with untreated and treated MSCs. One week after the last injection, mice sacrificed and samples were taken and the desired evaluations were done. RESULTS Our results revealed that the respiratory burst capacity, neutrophil phagocytosis, and nitric oxide production in the population of splenocytes were higher in the positive control group compared to the treatment groups. Also, the level of production of IL-4, IL-10 and TGF-β cytokines and INF-γ and IL-17 cytokines showed a significant increase and decrease, respectively, compared to the positive control group. CONCLUSION Treatment of MSCs with calcitriol leads to an improvement in regulatory function and inhibitory effects on inflammatory mediators of innate immune cells, particularly splenocytes, in a rheumatoid arthritis model compared to untreated mesenchymal stem cells.
Collapse
Affiliation(s)
- Alireza Rafati
- Department of Medical Genetics, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Reihaneh Ramezani
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hadi Esmaeili Gouvarchin Ghaleh
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shabnam Bahrami
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Mahmood Reza Masoudi
- School of Medical Sciences, Emam Reza Hospital Sirjan Faculty of Medical Sciences, Sirjan, Iran
| |
Collapse
|
8
|
Wu T, Li Y, Liu Y, Chu CQ. Preclinical RA: How to halt its progression. Best Pract Res Clin Rheumatol 2025; 39:102030. [PMID: 39721896 DOI: 10.1016/j.berh.2024.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 12/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder with a complex pathogenesis that evolves through various stages before clinical symptoms emerge. This review outlines the natural history of RA, starting from genetic predisposition and environmental triggers to preclinical autoimmunity and subsequent joint inflammation. Key genetic factors interact with environmental elements like smoking and infections, producing autoantibodies such as anti-citrullinated protein antibodies (ACPA) and rheumatoid factor, which precede clinical manifestations by several years. The preclinical phases offer critical opportunities for intervention aiming at halting disease progression. Preventive strategies including lifestyle modifications, dietary interventions, and targeted immune modulation may halt the progression to clinical RA in those at-risk individuals.
Collapse
Affiliation(s)
- Tong Wu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Lecheng Hospital, Sichuan University, Boao, Hainan, 571435, China.
| | - Cong-Qiu Chu
- Division of Arthritis and Rheumatic Diseases, Oregon Health & Science University, Portland, OR, 97239, USA; Rheumatology Section, VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
9
|
Pareek D, Zeyaullah M, Patra S, Alagu O, Singh G, Wasnik K, Gupta PS, Paik P. Mesoporous polymeric nanoparticles for effective treatment of inflammatory diseases: an in vivo study. J Mater Chem B 2025; 13:3094-3113. [PMID: 39902477 DOI: 10.1039/d4tb02012j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Acute inflammatory diseases require suitable medicine over the existing therapeutics. In this line, the present work is focused on developing polymeric nanomedicine for the treatment of inflammatory disorders. Herein, cell viable nanoparticles (GlyNPs) of size 180-250 nm in diameter and pore size of 4-5 nm in diameter, based on glycine and acryloyl chloride, have been developed and proved to be a potential anti-inflammatory agent without using any conventional drugs. These particles exhibit colloidal stability (with a zeta potential of -35.6 mV). A network pharmacology-based computational study has been executed on 9076 genes and proteins responsible for inflammatory diseases, out of which 10 are selected that have a major role in rheumatoid arthritis (RA). In silico docking study has been conducted to find out the targeted efficiency of the GlyNPs considering 10 inflammation-specific markers, namely IL-6, IL-1β, TNF-α, TLR-4, STAT-1, MAPK-8, MAPK-14, iNOS, NF-κβ and COX-2. The results revealed that the GlyNPs could be an excellent anti-inflammatory component similar to aspirin. The in vitro inflammation activity of these GlyNPs has also been checked on an inflammation model generated by LPS in RAW 264.7 macrophages. Then, the in vitro anti-inflammation efficiency has been checked with 10-150 μg mL-1 of GlyNP doses. The treatment efficiency has been checked on inflammation-responsible immune markers (NO level, NF-κβ, INF-γ, IL-6, IL-10, and TNF-α) and it was found that the GlyNPs are an excellent component in reducing inflammation. The in vivo therapeutic response of GlyNPs on the induced rheumatoid arthritis (RA) model has been evaluated by measuring the morphological, biochemical and immune-cytokine and interferon levels responsible for the inflammation, using a 2 g kg-1 dose (sample to weight of rat). The anti-inflammatory efficiency of GlyNPs without using additional drugs was found to be excellent. Thus, GlyNPs could be paramount for the potential treatment of various inflammatory diseases.
Collapse
Affiliation(s)
- Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Md Zeyaullah
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Oviya Alagu
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Gurmeet Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| |
Collapse
|
10
|
Cui P, Cheng T, Yan H, Xu D, Ren G, Ma S. The Value of NLR and PLR in the Diagnosis of Rheumatoid Arthritis Combined with Interstitial Lung Disease and Assessment of Treatment Effect: A Retrospective Cohort Study. Int J Gen Med 2025; 18:867-880. [PMID: 39990294 PMCID: PMC11847414 DOI: 10.2147/ijgm.s509546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Objective This retrospective cohort study investigated the value of neutrophil-lymphocyte ratio (NLR) and platelet-lymphocyte ratio (PLR) in the diagnosis and treatment of rheumatoid arthritis complicated with interstitial lung disease (RA-ILD). Methods A total of 163 patients with newly diagnosed rheumatoid arthritis (RA) were enrolled, with 122 patients in the RA group and 41 patients in the RA-ILD group. The mean age of the RA group was 63.84 ± 8.53 years, with a male-to-female ratio of 14:47. The RA-ILD group had a mean age of 66.29 ± 12.72 years, with a male-to-female ratio of 13:28. During the 2-year follow-up period, 10 patients in the RA group developed interstitial lung disease (ILD). Results NLR and PLR were significantly higher in RA-ILD group than in RA group (p < 0.05). The optimal critical values of NLR and PLR for the diagnosis of RA-ILD were 3.15 and 152.62, the area under ROC curve was 0.615 and 0.61, the sensitivity was 72%, 62%, and the specificity was 54% and 64%. NLR and PLR were significantly increased after ILD during follow-up in RA patients but decreased after ILD in the predicted percentage of vital capacity (VC%), forced vital capacity (FVC%), forced expiratory volume in the first second (FEV1%) and carbon monoxide dispersion (DLcoSB%) (p < 0.05). Moreover, NLR and PLR decreased after treatment. While VC%, FVC%, FEV1%, and DLcoSB% increased after treatment (p < 0.05). NLR was negatively correlated with FVC% and DLcoSB% both before and after treatment. PLR was also significantly negatively correlated with FVC% and DLcoSB% before and after treatment (p < 0.05). Conclusion When NLR and PLR increase, we should be alert to the possibility of RA complicated with ILD, which can be used as an evaluation index of the treatment effect of RA-ILD.
Collapse
Affiliation(s)
- Peng Cui
- Rheumatology Immunology Department, Zibo First Hospital, Zibo, People’s Republic of China
| | - Tao Cheng
- Rheumatology Immunology Department, Zibo First Hospital, Zibo, People’s Republic of China
| | - Huichun Yan
- Rheumatology Immunology Department, Zibo First Hospital, Zibo, People’s Republic of China
| | - Dong Xu
- Rheumatology Immunology Department, Zibo First Hospital, Zibo, People’s Republic of China
| | - Guohua Ren
- Rheumatology Immunology Department, Zibo First Hospital, Zibo, People’s Republic of China
| | - Shangmin Ma
- Rheumatology Immunology Department, Zibo First Hospital, Zibo, People’s Republic of China
| |
Collapse
|
11
|
Huang L, Shi J, Li H, Lin Q. Bidirectional Mendelian randomization reveals causal immune cell phenotypes in rheumatoid arthritis. Medicine (Baltimore) 2025; 104:e41512. [PMID: 39928790 PMCID: PMC11813058 DOI: 10.1097/md.0000000000041512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/12/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by persistent inflammation in synovial joints and influenced by genetic and environmental factors. The immune system, comprising various cells that either exacerbate or reduce inflammation, plays a central role in RA pathogenesis. Despite extensive research, the causal relationships between immune cell phenotypes and RA remain unclear. We employed a two-sample Mendelian randomization (MR) approach to investigate the causal associations between 731 immune cell phenotypes and RA. Genetic variants, carefully selected based on rigorous criteria, served as instrumental variables to ensure analytical validity. Data on RA were derived from the FinnGen database, whereas immune cell phenotype data were obtained from the genome-wide association studies catalog. We employed 5 MR methods, including inverse variance weighted and MR Egger, to ensure robust causal inference. We assessed for pleiotropy and heterogeneity and adjusted findings using the False Discovery Rate. After False Discovery Rate adjustment (threshold < 0.05), inverse variance weighted analysis revealed potential causal relationships between 4 immune cell types and RA: CD62L- dendritic cells %dendritic cells (P = 3.88E-05; 95% confidence interval [CI] = 1.056), CD19 on IgD+ CD38- naive cells (P = 1.75E-04; 95% CI = 0.969), CD45RA on TD CD8br (P = 5.59E-04; 95% CI = 0.919), and HLA DR on CD33- HLA DR+ (P = 8.13E-05; 95% CI = 1.422). In reverse Mendelian studies, specific immune cell phenotypes were found to be associated with RA risk and progression: the percentage of memory B cells among lymphocytes (P = 2.74E-04; 95% CI = 0.861), IgD+ CD24+ cells among lymphocytes (P = 6.93E-04; 95% CI = 0.867), CD4+ CD8dim cells among lymphocytes (P = 2.92E-04; 95% CI = 0.802), CD4+ CD8dim cells among leukocytes (P = 4.37E-04; 95% CI = 0.814), and CD24 expression on IgD+CD24+ cells (P = 1.05E-04; 95% CI = 0.857). These results identify immune cell phenotypes closely linked to RA susceptibility and progression. The findings suggest that specific immune cell phenotypes are not only influenced by RA but may also contribute to its development and progression. These results offer new insights into the immunological underpinnings of RA and highlight potential targets for therapeutic intervention. Future research should focus on validating these causal relationships in diverse populations and exploring the molecular mechanisms involved.
Collapse
Affiliation(s)
- Luofei Huang
- Liuzhou Municipal Liutie Central Hospital, Liuzhou, Guangxi, China
| | - Jian Shi
- Department of Internal Medicine, The People’s Hospital of Laibin, Laibin, Guangxi, China
| | - Han Li
- Department of Internal Medicine, Liuzhou People’s Hospital, Liuzhou, Guangxi, China
| | - Quanzhi Lin
- Department of Internal Medicine, The First Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| |
Collapse
|
12
|
Thumsi A, Martínez D, Swaminathan SJ, Esrafili A, Suresh AP, Jaggarappu MMC, Lintecum K, Halim M, Mantri SV, Sleiman Y, Appel N, Gu H, Curtis M, Zuniga C, Acharya AP. Inverse-Vaccines for Rheumatoid Arthritis Re-establish Metabolic and Immunological Homeostasis in Joint Tissues. Adv Healthc Mater 2025; 14:e2303995. [PMID: 38469995 PMCID: PMC11390975 DOI: 10.1002/adhm.202303995] [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: 11/14/2023] [Revised: 03/06/2024] [Indexed: 03/13/2024]
Abstract
Rheumatoid arthritis (RA) causes immunological and metabolic imbalances in tissue, exacerbating inflammation in affected joints. Changes in immunological and metabolic tissue homeostasis at different stages of RA are not well understood. Herein, the changes in the immunological and metabolic profiles in different stages in collagen induced arthritis (CIA), namely, early, intermediate, and late stage is examined. Moreover, the efficacy of the inverse-vaccine, paKG(PFK15+bc2) microparticle, to restore tissue homeostasis at different stages is also investigated. Immunological analyses of inverse-vaccine-treated group revealed a significant decrease in the activation of pro-inflammatory immune cells and remarkable increase in regulatory T-cell populations in the intermediate and late stages compared to no treatment. Also, glycolysis in the spleen is normalized in the late stages of CIA in inverse-vaccine-treated mice, which is similar to no-disease tissues. Metabolomics analyses revealed that metabolites UDP-glucuronic acid and L-Glutathione oxidized are significantly altered between treatment groups, and thus might provide new druggable targets for RA treatment. Flux metabolic modeling identified amino acid and carnitine pathways as the central pathways affected in arthritic tissue with CIA progression. Overall, this study shows that the inverse-vaccines initiate early re-establishment of homeostasis, which persists through the disease span.
Collapse
Affiliation(s)
- Abhirami Thumsi
- The Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Diego Martínez
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | | | - Arezoo Esrafili
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Abhirami P. Suresh
- The Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | | | - Kelly Lintecum
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Michelle Halim
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Shivani V. Mantri
- Department of Biomedical Engineering, School of Biological and Health System Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Yasmine Sleiman
- Department of Biomedical Engineering, School of Biological and Health System Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Nicole Appel
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ, 85281, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA, 85281
| | - Marion Curtis
- Department of Cancer Biology, Mayo Clinic, Scottsdale, AZ, 85259 8, USA
- College of Medicine and Science, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Cristal Zuniga
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | - Abhinav P. Acharya
- The Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, 44106, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, 44106, USA
| |
Collapse
|
13
|
Wang X, Ni T, Miao J, Huang X, Feng Z. The role and mechanism of triptolide, a potential new DMARD, in the treatment of rheumatoid arthritis. Ageing Res Rev 2025; 104:102643. [PMID: 39722411 DOI: 10.1016/j.arr.2024.102643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Triptolide (TP) is the primary pharmacological component of Tripterygium Glycosides (TG), which has anti-inflammatory, antiproliferative, and immunosuppressive properties, among other pharmacological actions, and has excellent potential for developing into a new DMARD. We have reviewed the effects and mechanisms of TP on immunosuppression, inhibiting synovial proliferation, and preventing articular bone destruction in the treatment of rheumatoid arthritis (RA), which is a common disease in the elderly in this paper. We have found that TP has regulatory effects on multiple vital cells in the above-mentioned pathological process of RA, such as monocytes/macrophages, dendritic cells, T cells, fibroblast-like synoviocytes, and osteoclasts. We also found that TP can regulate multiple key signaling pathways such as NF-κB, JAK/STAT, and MAPK through various molecular regulatory mechanisms, achieving regulatory effects on numerous phenotypes of the above-mentioned vital cells.
Collapse
Affiliation(s)
- Xiwen Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Tianyang Ni
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Jianru Miao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Xinyao Huang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Zhe Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China.
| |
Collapse
|
14
|
Ding C, Liu Q, You X, Yuan J, Xia J, Tan Y, Hu Y, Wang Q. Investigating the molecular mechanism of epimedium herb in treating rheumatoid arthritis through network pharmacology, molecular docking, and experimental validation. Mol Divers 2025:10.1007/s11030-024-11019-z. [PMID: 39821498 DOI: 10.1007/s11030-024-11019-z] [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: 09/09/2024] [Accepted: 10/13/2024] [Indexed: 01/19/2025]
Abstract
This study attempted to explore the molecular mechanism of Epimedium herb (EH) on rheumatoid arthritis (RA) treatment. We employed network pharmacology, molecular docking, and HPLC analysis to investigate the molecular mechanisms underlying the efficacy of EH in treating RA. To assess the efficacy of EH intervention, RA fibroblast-like synoviocytes (RA-FLS) and collagen-induced arthritis (CIA) mouse models were utilized. Ultimately, the active compounds icariin, luteolin, quercetin, and kaempferol were identified, with interleukin-1β (IL-1β), IL-6, tumor necrosis factor-alpha (TNF-α), and matrix metalloproteinase-9 (MMP-9) emerging as key targets of EH for RA. These targets were found to be downregulated in both in vitro and in vivo experiments following EH intervention. Furthermore, EH treatment induced apoptosis, reduced metastasis and invasion in RA-FLS, and ameliorated arthritis-related symptoms while regulating Th17 and Treg cells in CIA mice.
Collapse
Affiliation(s)
- Chunhui Ding
- Department of Pharmacy, Wuxi Ninth People's Hospital Affiliated to Soochow University, No. 999 Liang Xi Road, Binhu District, Wuxi, 214000, Jiangsu, China
| | - Qingyang Liu
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214000, Jiangsu, China
| | - Xiaohong You
- Department of Pharmacy, Wuxi Ninth People's Hospital Affiliated to Soochow University, No. 999 Liang Xi Road, Binhu District, Wuxi, 214000, Jiangsu, China
| | - Jianming Yuan
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214000, Jiangsu, China
| | - Jinjun Xia
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214000, Jiangsu, China
| | - Yuan Tan
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214000, Jiangsu, China
| | - Yunxia Hu
- Department of Rheumatology and Immunology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214000, Jiangsu, China.
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, 214000, Jiangsu, China.
| |
Collapse
|
15
|
Jiang Y, Hu Z, Huang R, Ho K, Wang P, Kang J. Metabolic reprogramming and macrophage expansion define ACPA-negative rheumatoid arthritis: insights from single-cell RNA sequencing. Front Immunol 2025; 15:1512483. [PMID: 39830504 PMCID: PMC11739280 DOI: 10.3389/fimmu.2024.1512483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/26/2024] [Indexed: 01/22/2025] Open
Abstract
Background Anti-citrullinated peptide antibodies (ACPA)-negative (ACPA-) rheumatoid arthritis (RA) presents significant diagnostic and therapeutic challenges due to the absence of specific biomarkers, underscoring the need to elucidate its distinctive cellular and metabolic profiles for more targeted interventions. Methods Single-cell RNA sequencing data from peripheral blood mononuclear cells (PBMCs) and synovial tissues of patients with ACPA- and ACPA+ RA, as well as healthy controls, were analyzed. Immune cell populations were classified based on clustering and marker gene expression, with pseudotime trajectory analysis, weighted gene co-expression network analysis (WGCNA), and transcription factor network inference providing further insights. Cell-cell communication was explored using CellChat and MEBOCOST, while scFEA enabled metabolic flux estimation. A neural network model incorporating key genes was constructed to differentiate patients with ACPA- RA from healthy controls. Results Patients with ACPA- RA demonstrated a pronounced increase in classical monocytes in PBMCs and C1QChigh macrophages (p < 0.001 and p < 0.05). Synovial macrophages exhibited increased heterogeneity and were enriched in distinct metabolic pathways, including complement cascades and glutathione metabolism. The neural network model achieved reliable differentiation between patients with ACPA- RA and healthy controls (AUC = 0.81). CellChat analysis identified CD45 and CCL5 as key pathways facilitating macrophage-monocyte interactions in ACPA- RA, prominently involving iron-mediated metabolite communication. Metabolic flux analysis indicated elevated beta-alanine and glutathione metabolism in ACPA- RA macrophages. Conclusion These findings underscore that ACPA-negative rheumatoid arthritis is marked by elevated classical monocytes in circulation and metabolic reprogramming of synovial macrophages, particularly in complement cascade and glutathione metabolism pathways. By integrating single-cell RNA sequencing with machine learning, this study established a neural network model that robustly differentiates patients with ACPA- RA from healthy controls, highlighting promising diagnostic biomarkers and therapeutic targets centered on immune cell metabolism.
Collapse
Affiliation(s)
- Yafeng Jiang
- Department of Hematology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhaolan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Roujie Huang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetric & Gynecologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kaying Ho
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
| | - Pengfei Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin Kang
- Department of Rheumatology and Immunology, the Second Xiangya Hospital of Central South University, Changsha, China
- Department of Rheumatology and Immunology, Clinical Medical Research Center for Systemic Autoimmune Diseases in Hunan Province, Changsha, China
| |
Collapse
|
16
|
Kim HR, Lee H, Kim TH, Gil M, Kim DW. Natural killer cell activity and its relationship with disease activity in rheumatoid arthritis patients. Hum Immunol 2025; 86:111185. [PMID: 39626408 DOI: 10.1016/j.humimm.2024.111185] [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: 03/27/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 01/25/2025]
Abstract
The aim of this study was to explore the relationship between natural killer cell activity (NKA) and disease activity of rheumatoid arthritis (RA). We retrospectively reviewed 259 patients' data including RA markers associated with disease activity and NKA measured by a blood NKA test. Patients were divided into two groups based on their NKA levels, a low NKA group (NKA < 100 pg/mL) and a high NKA group (NKA 100-250 pg/mL). The low NKA group exhibited heightened RA characteristics, including increased seropositivity, anti-cyclic citrullinated peptide (anti-CCP) antibodies, erythrocyte sedimentation rate (ESR), 28-joint disease activity score, tender joints, visual analog scale (VAS), and TNF-α antagonist usage. A negative correlation was observed between NKA and RA severity metrics, including rheumatoid factor (RF), anti-CCP antibodies, ESR, C-reactive protein, tender and swollen joints, and VAS scores. Logistic regression analysis indicated that factors such as seropositivity, elevated RF and anti-CCP antibodies, increased tender and swollen joints, higher VAS scores, and the employment of biological agents were linked with higher chances of belonging to the lower NKA group. Comparable trends were found within the seropositive RA patient subset. Our findings highlight a significant link between diminished NKA levels and exacerbated RA symptoms.
Collapse
Affiliation(s)
- Hye Rim Kim
- NKMAX Co., Ltd., Seongnam, Republic of Korea
| | - Hyeja Lee
- NKMAX Co., Ltd., Seongnam, Republic of Korea
| | - Tae Hee Kim
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Minchan Gil
- NKMAX Co., Ltd., Seongnam, Republic of Korea.
| | - Dong Wook Kim
- Department of Internal Medicine, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
17
|
Quah S, Subramanian G, Tan JSL, Utami KH, Sampath P. MicroRNAs: a symphony orchestrating evolution and disease dynamics. Trends Mol Med 2025; 31:21-35. [PMID: 39112313 DOI: 10.1016/j.molmed.2024.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/13/2024] [Accepted: 07/08/2024] [Indexed: 01/11/2025]
Abstract
The genesis of human disease lies in our evolutionary past. Evolution has featured a general trend towards increased morphological complexity, partly conferred by expansion in gene regulatory capacity via microRNA (miRNA) innovation. Many human diseases are directly related to the evolved roles of these miRNAs, and miRNA-based therapies are emerging as an appealing strategy for precision medicine. We focus on three categories of human disease - cancer, inflammation-linked pathologies, and neurological disorders - which are highly prevalent and are associated with substantial disease burden worldwide. In each category we discuss the pathogenic roles of miRNAs in the context of their evolved functions, as well as current and potential advances in targeting these miRNAs for disease therapy.
Collapse
Affiliation(s)
- Shan Quah
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Republic of Singapore
| | - Gowtham Subramanian
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Republic of Singapore
| | - Jonathan S L Tan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Republic of Singapore
| | | | - Prabha Sampath
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove #06-06 Immunos, Singapore 138648, Republic of Singapore; Genome Institute of Singapore, Agency for Science, Technology, and Research, 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore; Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Republic of Singapore; Skin Research Institute of Singapore (SRIS), 11 Mandalay Road #17-01 Clinical Sciences Building, Singapore 308232, Republic of Singapore.
| |
Collapse
|
18
|
Wang J, Zhu R, Fang H, Xing X, Ge L, Cai G. Association of prognostic nutritional index with the presence and all-cause mortality of rheumatoid arthritis: the National Health and Nutrition Examination Survey 2003-2018. BMC Public Health 2024; 24:3281. [PMID: 39593001 PMCID: PMC11590335 DOI: 10.1186/s12889-024-20795-0] [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: 04/21/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The prognostic nutritional index (PNI) is a comprehensive measure of individual immune and nutritional status. This study aimed to evaluate the role of PNI in the presence and mortality of rheumatoid arthritis (RA). METHODS This study used data of participants aged ≥ 40 years from the National Health and Nutrition Examination Survey (NHANES) 2003-2018. PNI was calculated using serum albumin and lymphocyte count. The relationship between PNI and the prevalence of RA and mortality among RA patients was assessed using logistic and Cox regression models. Nonlinear associations were explored using restricted cubic splines (RCS). RESULTS Of 18,245 participants (mean 55.4 years, 49% female), 1901 had RA, among whom (480/1899, 25%) died during a median follow-up period of 84 months. PNI was inversely associated with the likelihood of having RA (odds ratio = 0.97, 95% confidence interval [CI]: 0.95-0.98). Compared to participants whose PNI was in the lowest quartile, those in other quartiles had a reduced likelihood of having RA by 21-38% (Ptrend <0.01). Cox regression analysis revealed an inverse association between PNI and all-cause mortality (hazard ratio = 0.95, 95%CI: 0.91-0.99). An L-shaped association was observed between PNI and the presence and all-cause mortality of RA, with turning points occurring around the mean value of PNI. The presence and all-cause mortality of RA was significantly reduced before the turning points of PNI and plateaued afterwards. CONCLUSION In middle-aged and older adults, there is an inverse association between PNI and the presence and all-cause mortality of RA.
Collapse
Affiliation(s)
- Junjie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Rui Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Haonan Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xing Xing
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Liru Ge
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Guoqi Cai
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, 230032, China.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, 7000, Australia.
| |
Collapse
|
19
|
Figueiredo ML. Applications of single-cell RNA sequencing in rheumatoid arthritis. Front Immunol 2024; 15:1491318. [PMID: 39600707 PMCID: PMC11588722 DOI: 10.3389/fimmu.2024.1491318] [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: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Single cell RNA sequencing (scRNA-seq) is a relatively new technology that provides an unprecedented, detailed view of cellular heterogeneity and function by delineating the transcriptomic difference among individual cells. This will allow for mapping of cell-type-specific signaling during physiological and pathological processes, to build highly specific models of cellular signaling networks between the many discrete clusters that are present. This technology therefore provides a powerful approach to dissecting the cellular and molecular mechanisms that contribute to autoimmune diseases, including rheumatoid arthritis (RA). scRNA-seq can offer valuable insights into RA unique cellular states and transitions, potentially enabling development of novel drug targets. However, some challenges that still limit its mainstream utilization and include higher costs, a lower sensitivity for low-abundance transcripts, and a relatively complex data analysis workflow relative to bulk or traditional RNA-seq. This minireview explores the emerging application of scRNA-seq in RA research, highlighting its role in producing important insights that can help pave the way for innovative and more effective therapeutic strategies.
Collapse
Affiliation(s)
- Marxa L. Figueiredo
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue
University, West Lafayette, IN, United States
| |
Collapse
|
20
|
Bufan B, Marčetić M, Djuretić J, Ćuruvija I, Blagojević V, Božić DD, Milutinović V, Janković R, Sopta J, Kotur-Stevuljević J, Arsenović-Ranin N. Evaluation of the Anti-Inflammatory/Immunomodulatory Effect of Teucrium montanum L. Extract in Collagen-Induced Arthritis in Rats. BIOLOGY 2024; 13:818. [PMID: 39452128 PMCID: PMC11505313 DOI: 10.3390/biology13100818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
The anti-inflammatory/immunomodulatory effects of Teucrium montanum L. (TM), a plant distributed in the Mediterranean region, have been insufficiently examined. The effects of the TM ethanol extract were tested in a rat collagen-induced arthritis (CIA) model of rheumatoid arthritis. LC-MS was used for the phytochemical analysis of the TM extract. Dark Agouti rats were immunized with bovine type II collagen (CII) in incomplete Freund's adjuvant for CIA, and treated with 100 or 200 mg/kg of TM extract daily via oral administration. Clinical and histopathological evaluations and a flow cytometric analysis of the phenotypic and functional characteristics of splenocytes and draining lymph node cells were performed. The cytokines in the paw tissue culture supernatants and anti-CII antibodies in serum were determined by ELISA. The TM extract, with the dominant components verbascoside and luteolin 7-O-rutinoside, reduced the arthritic score and ankle joint inflammation in CIA rats, promoted the antioxidant profile in serum, and lowered pro-inflammatory TNF-α, IL-6 and IL-1β production. It suppressed the activation status of CD11b+ cells by lowering CD86, MHCII and TLR-4 expression, and promoted the Th17/T regulatory cell (Tregs) balance towards Tregs. A lower frequency of B cells was accompanied by a lower level of anti-CII antibodies in treated rats. These findings imply the favorable effect of TM extract on the clinical presentation of CIA, suggesting its anti-inflammatory/immunomodulatory action and potential therapeutic effect.
Collapse
Affiliation(s)
- Biljana Bufan
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (B.B.); (D.D.B.)
| | - Mirjana Marčetić
- Department of Pharmacognosy, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (M.M.); (V.M.)
| | - Jasmina Djuretić
- Department of Pathobiology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia;
| | - Ivana Ćuruvija
- Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ć.); (V.B.)
| | - Veljko Blagojević
- Institute of Virology, Vaccines and Sera “Torlak”, 11221 Belgrade, Serbia; (I.Ć.); (V.B.)
| | - Dragana D. Božić
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (B.B.); (D.D.B.)
| | - Violeta Milutinović
- Department of Pharmacognosy, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (M.M.); (V.M.)
| | - Radmila Janković
- Institute of Pathology “Prof. dr Đorđe Joannović”, University of Belgrade-Faculty of Medicine, 11000 Belgrade, Serbia; (R.J.); (J.S.)
| | - Jelena Sopta
- Institute of Pathology “Prof. dr Đorđe Joannović”, University of Belgrade-Faculty of Medicine, 11000 Belgrade, Serbia; (R.J.); (J.S.)
| | - Jelena Kotur-Stevuljević
- Department of Medical Biochemistry, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia;
| | - Nevena Arsenović-Ranin
- Department of Microbiology and Immunology, University of Belgrade-Faculty of Pharmacy, 11221 Belgrade, Serbia; (B.B.); (D.D.B.)
| |
Collapse
|
21
|
DERVISEVIC A, FAJKIC A, JAHIC E, DERVISEVIC L, AJANOVIC Z, ADEMOVIC E, ZACIRAGIC A. Systemic Immune-inflammation Index in Evaluation of Inflammation in Rheumatoid Arthritis Patients. Medeni Med J 2024; 39:183-191. [PMID: 39350540 PMCID: PMC11572210 DOI: 10.4274/mmj.galenos.2024.60533] [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: 03/22/2024] [Accepted: 07/31/2024] [Indexed: 11/20/2024] Open
Abstract
Objective To evaluate the systemic immune-inflammation (SII) index in patients with rheumatoid arthritis (RA) stratified by systemic inflammatory status. Methods Seropositive patients with RA (n=58) were divided into two groups based on serum hs-C-reactive protein (hs-CRP) levels: RA patients with hs-CRP levels of at or 3 mg/L or above (high systemic inflammatory status; n=38) and RA patients with hs-CRP levels of less than 3 mg/L (low systemic inflammatory status; n=20). The control group comprised 31 healthy individuals. Blood samples were tested for the next parameters: leukocytes, neutrophilic granulocytes, lymphocytes, thrombocytes [platelet (PLT)], high-sensitivity hs-CRP, sed rate [erythrocyte sedimentation rate (ESR)], neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and monocyte-to-lymphocyte ratio (MLR). The SII index was derived as Neu x PLT/Lym. Results In patients with RA, the SII index was elevated compared with that of healthy individuals and positively correlated with hs-CRP, erythrocyte sedimentation rate, NLR, MLR, PLR, tender joint count, and swollen-to-tender joint count ratio. Patients with RA who had hs-CRP levels of 3 mg/L above exhibited a statistically significant increase in the SII compared with those with hs-CRP levels below 3 mg/L. Additionally, within the cohort of RA patients with hs-CRP levels at or above 3 mg/L, a positive correlation was found between the SII index and both NLR and PLR. The SII index was positively correlated with NLR, MLR, and PLR in RA patients with hs-CRP levels below 3 mg/L. The cut-off point of the SII index for distinguishing between RA cases with hs-CRP levels 3 mg/L and those with hs-CRP levels 3 mg/L or higher was ≥323.4, with a sensitivity of 77.6% and a specificity of 54.8%. Conclusions The serum SII index can be a potentially useful marker for evaluating the inflammatory process and clinical progression of RA.
Collapse
Affiliation(s)
- Amela DERVISEVIC
- University of Sarajevo Faculty of Medicine, Department of Human Physiology, Sarajevo, Bosnia and Herzegovina
| | - Almir FAJKIC
- University of Sarajevo Faculty of Medicine, Department of Pathophysiology, Sarajevo, Bosnia and Herzegovina
| | - Elmedina JAHIC
- Clinical Center University of Sarajevo, Clinic for Heart, Blood Vessel and Rheumatic Diseases, Sarajevo, Bosnia and Herzegovina
| | - Lejla DERVISEVIC
- University of Sarajevo Faculty of Medicine, Department of Anatomy, Sarajevo, Bosnia and Herzegovina
| | - Zurifa AJANOVIC
- University of Sarajevo Faculty of Medicine, Department of Anatomy, Sarajevo, Bosnia and Herzegovina
| | - Enisa ADEMOVIC
- University of Sarajevo Faculty of Medicine, Department of Epidemiology and Biostatistics, Sarajevo, Bosnia and Herzegovina
| | - Asija ZACIRAGIC
- University of Sarajevo Faculty of Medicine, Department of Human Physiology, Sarajevo, Bosnia and Herzegovina
| |
Collapse
|
22
|
Yin X, Wang Q, Tang Y, Wang T, Zhang Y, Yu T. Research progress on macrophage polarization during osteoarthritis disease progression: a review. J Orthop Surg Res 2024; 19:584. [PMID: 39342341 PMCID: PMC11437810 DOI: 10.1186/s13018-024-05052-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/01/2024] [Indexed: 10/01/2024] Open
Abstract
Primary osteoarthritis (OA) is a prevalent degenerative joint disease that mostly affects the knee joint. It is a condition that occurs around the world. Because of the aging population and the increase in obesity prevalence, the incidence of primary OA is increasing each year. Joint replacement can completely subside the pain and minimize movement disorders caused by advanced OA, while nonsteroidal drugs and injection of sodium hyaluronate into the joint cavity can only partially relieve the pain; hence, it is critical to search for new methods to treat OA. Increasing lines of evidence show that primary OA is a chronic inflammatory disorder, with synovial inflammation as the main characteristic. Macrophages, as one of the immune cells, can be polarized to produce M1 (proinflammatory) and M2 (anti-inflammatory) types during synovial inflammation in OA. Following polarization, macrophages do not come in direct contact with chondrocytes; however, they affect chondrocyte metabolism through paracrine production of a significant quantity of inflammatory cytokines, matrix metalloproteinases, and growth factors and thus participate in inducing joint pain, cartilage injury, angiogenesis, and osteophyte formation. The main pathways that influence the polarization of macrophages are the Toll-like receptor and NF-κB pathways. The study of how macrophage polarization affects OA disease progression has gradually become one of the approaches to prevent and treat OA. Experimental studies have found that the treatment of macrophage polarization in primary OA can effectively relieve synovial inflammation and reduce cartilage damage. The present article summarizes the influence of inflammatory factors secreted by macrophages after polarization on OA disease progression, the main signaling pathways that induce macrophage differentiation, and the role of different polarized types of macrophages in OA; thus, providing a reference for preventing and treating primary OA.
Collapse
Affiliation(s)
- Xiangzhi Yin
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
- Department of Orthopaedics, Qingdao Municipal Hospital, Qingdao, 266011, China
| | - Quan Wang
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266005, China
| | - Yijie Tang
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Tianrui Wang
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Yingze Zhang
- Department of Orthopaedics, Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Tengbo Yu
- Department of Orthopaedics, Qingdao Municipal Hospital, Qingdao, 266011, China.
| |
Collapse
|
23
|
Zhu Q, Zhou H. The role of cGAS-STING signaling in rheumatoid arthritis: from pathogenesis to therapeutic targets. Front Immunol 2024; 15:1466023. [PMID: 39386207 PMCID: PMC11461283 DOI: 10.3389/fimmu.2024.1466023] [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: 07/17/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease primarily characterized by erosive and symmetric polyarthritis. As a pivotal axis in the regulation of type I interferon (IFN-I) and innate immunity, the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway has been implicated in the pathogenesis of RA. This pathway mainly functions by regulating cell survival, pyroptosis, migration, and invasion. Therefore, understanding the sources of cell-free DNA and the mechanisms underlying the activation and regulation of cGAS-STING signaling in RA offers a promising avenue for targeted therapies. Early detection and interventions targeting the cGAS-STING signaling are important for reducing the medical burden on individuals and healthcare systems. Herein, we review the existing literature pertaining to the role of cGAS-STING signaling in RA, and discuss current applications and future directions for targeting the cGAS-STING signaling in RA treatments.
Collapse
Affiliation(s)
- Qiugang Zhu
- Department of Laboratory Medicine, Shangyu People’s Hospital of Shaoxing, Shaoxing University, Shaoxing, China
| | - Huimin Zhou
- Department of Laboratory Medicine, Wuxi Ninth People’s Hospital Affiliated to Soochow University, Wuxi, China
| |
Collapse
|
24
|
Zhou R, Xue S, Cheng Y, Chen Y, Wang Y, Xing J, Liu H, Xu Y, Lin Y, Pei Z, Wei X, Ding J, Li S, Wang K, Yao F, Zhao Y, Ding C, Hu W. Macrophage membrane-camouflaged biomimetic nanoparticles for rheumatoid arthritis treatment via modulating macrophage polarization. J Nanobiotechnology 2024; 22:578. [PMID: 39300463 DOI: 10.1186/s12951-024-02822-9] [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: 05/26/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024] Open
Abstract
Rheumatoid arthritis (RA) is a debilitating autoimmune disease characterized by chronic joint inflammation and cartilage damage. Current therapeutic strategies often result in side effects, necessitating the development of targeted and safer treatment options. This study introduces a novel nanotherapeutic system, 2-APB@DGP-MM, which utilizes macrophage membrane (MM)-encapsulated nanoparticles (NPs) for the targeted delivery of 2-Aminoethyl diphenylborinate (2-APB) to inflamed joints more effectively. The NPs are designed with a matrix metalloproteinase (MMP)-cleavable peptide, allowing for MMP-responsive drug release within RA microenvironment. Comprehensive in vitro and in vivo assays confirmed the successful synthesis and loading of 2-APB into the DSPE-GPLGVRGC-PEG (DGP) NPs, as well as their ability to repolarize macrophages from a pro-inflammatory M1 to an anti-inflammatory M2 phenotype. The NPs demonstrated high biocompatibility, low cytotoxicity, and enhanced cellular uptake. In a collagen-induced arthritis (CIA) mouse model, intra-articular injection of 2-APB@DGP-MM significantly reduced synovial inflammation and cartilage destruction. Histological analysis corroborated these findings, demonstrating marked improvements in joint structure and delayed disease progression. Above all, the 2-APB@DGP-MM nanotherapeutic system offers a promising and safe approach for RA treatment by modulating macrophage polarization and delivering effective agents to inflamed joints.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Song Xue
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510200, China
| | - Yuanzhi Cheng
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yan Wang
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Jing Xing
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Hao Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Yucai Xu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Yi Lin
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Zejun Pei
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Xin Wei
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, People's Republic of China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Feng Yao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510200, China.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
25
|
Kumar A, Tatarian J, Shakhnovich V, Chevalier RL, Sudman M, Lovell DJ, Thompson SD, Becker ML, Funk RS. Identification of Plasma Metabolomic Biomarkers of Juvenile Idiopathic Arthritis. Metabolites 2024; 14:499. [PMID: 39330506 PMCID: PMC11434325 DOI: 10.3390/metabo14090499] [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: 07/29/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
Identification of disease and therapeutic biomarkers remains a significant challenge in the early diagnosis and effective treatment of juvenile idiopathic arthritis (JIA). In this study, plasma metabolomic profiling was conducted to identify disease-related metabolic biomarkers associated with JIA. Plasma samples from treatment-naïve JIA patients and non-JIA reference patients underwent global metabolomic profiling across discovery (60 JIA, 60 non-JIA) and replication (49 JIA, 38 non-JIA) cohorts. Univariate analysis identified significant metabolites (q-value ≤ 0.05), followed by enrichment analysis using ChemRICH and metabolic network mapping with MetaMapp and Cytoscape. Receiver operating characteristic (ROC) analysis determined the top discriminating biomarkers based on area under the curve (AUC) values. A total of over 800 metabolites were measured, consisting of 714 known and 155 unknown compounds. In the discovery cohort, 587 metabolites were significantly altered in JIA patients compared with the reference population (q < 0.05). In the replication cohort, 288 metabolites were significantly altered, with 78 overlapping metabolites demonstrating the same directional change in both cohorts. JIA was associated with a notable increase in plasma levels of sphingosine metabolites and fatty acid ethanolamides and decreased plasma levels of sarcosine, iminodiacetate, and the unknown metabolite X-12462. Chemical enrichment analysis identified cycloparaffins in the form of naproxen and its metabolites, unsaturated lysophospholipids, saturated phosphatidylcholines, sphingomyelins, ethanolamines, and saturated ceramides as the top discriminating biochemical clusters. ROC curve analysis identified 11 metabolites classified as highly discriminatory based on an AUC > 0.90, with the top discriminating metabolite being sphinganine-1-phosphate (AUC = 0.98). This study identifies specific metabolic changes in JIA, particularly within sphingosine metabolism, through both discovery and replication cohorts. Plasma metabolomic profiling shows promise in pinpointing JIA-specific biomarkers, differentiating them from those in healthy controls and Crohn's disease, which may improve diagnosis and treatment.
Collapse
Affiliation(s)
- Amar Kumar
- Center for Computational Biology, University of Kansas, Lawrence, KS 66047, USA
| | - Joshua Tatarian
- University of Kansas School of Medicine, Kansas City, KS 66160, USA
| | | | - Rachel L Chevalier
- University of Missouri-Kansas City School of Medicine & Children's Mercy Kansas City, Kansas City, MO 64108, USA
| | - Marc Sudman
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Daniel J Lovell
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Susan D Thompson
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mara L Becker
- Division of Rheumatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ryan S Funk
- Center for Computational Biology, University of Kansas, Lawrence, KS 66047, USA
- University of Kansas School of Medicine, Kansas City, KS 66160, USA
| |
Collapse
|
26
|
Matsumoto T, Nagano T, Taguchi K, Kobayashi T, Tanaka-Totoribe N. Toll-like receptor 3 involvement in vascular function. Eur J Pharmacol 2024; 979:176842. [PMID: 39033837 DOI: 10.1016/j.ejphar.2024.176842] [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: 05/12/2024] [Revised: 06/24/2024] [Accepted: 07/19/2024] [Indexed: 07/23/2024]
Abstract
Maintaining endothelial cell (EC) and vascular smooth muscle cell (VSMC) integrity is an important component of human health and disease because both EC and VSMC regulate various functions, including vascular tone control, cellular adhesion, homeostasis and thrombosis regulation, proliferation, and vascular inflammation. Diverse stressors affect functions in both ECs and VSMCs and abnormalities of functions in these cells play a crucial role in cardiovascular disease initiation and progression. Toll-like receptors (TLRs) are important detectors of pathogen-associated molecular patterns derived from various microbes and viruses as well as damage-associated molecular patterns derived from damaged cells and perform innate immune responses. Among TLRs, several studies reveal that TLR3 plays a key role in initiation, development and/or protection of diseases, and an emerging body of evidence indicates that TLR3 presents components of the vasculature, including ECs and VSMCs, and plays a functional role. An agonist of TLR3, polyinosinic-polycytidylic acid [poly (I:C)], affects ECs, including cell death, inflammation, chemoattractant, adhesion, permeability, and hemostasis. Poly (I:C) also affects VSMCs including inflammation, proliferation, and modulation of vascular tone. Moreover, alterations of vascular function induced by certain molecules and/or interventions are exerted through TLR3 signaling. Hence, we present the association between TLR3 and vascular function according to the latest studies.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan.
| | - Takayuki Nagano
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Naoko Tanaka-Totoribe
- First Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan
| |
Collapse
|
27
|
Elsayed Ramadan Genena S, Hamouda MAF, Salama NM, Zahran ES, Abdel Latif AA, Dawood AA. Interferon-induced protein 44 ( IFI44) and interferon regulatory factor 4 ( IRF4) gene expression in rheumatoid arthritis. J Immunoassay Immunochem 2024; 45:432-451. [PMID: 39051937 DOI: 10.1080/15321819.2024.2381524] [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] [Indexed: 07/27/2024]
Abstract
BACKGROUND AND OBJECTIVES The type I interferon (IFN) signature has been found to be overactivated in many systemic autoimmune diseases. This may be explained by impaired regulation of interferon-stimulated genes (ISGs) as well as interferon-induced protein 44 (IFI44) expression via their regulatory mechanisms via interferon regulatory factors (IRFs). PATIENTS AND METHODS This case-control study includes two groups: 50 RA patients and 50 healthy controls. The quantification of IFI44 and IRF4 expression levels by the real-time PCR technique was estimated. Disease Activity Score-28 (DAS-28) was estimated for RA patients only. RESULTS Among the RA patients, there were statistically significant increased ESR, CRP, TLC, RF, and anti-CCP levels (p value < 0.001) and significant increased expression of the IFI44 and IRF4 genes (p value < 0.001). There was a significant positive correlation between the IFI44 and IRF4, and there was a significant correlation between both and ESR and anti-CCP among RA patients. At a cutoff point of 1.95, IFI44 shows higher sensitivity and specificity values than IRF4 for the diagnosis of RA. CONCLUSION IFI44 was more sensitive for RA diagnosis than IRF4. IFI44 and IRF4 overexpression could be promising predictors of RA diagnosis and might become useful clinical tools to guide therapeutic strategies.
Collapse
Affiliation(s)
- Shaimaa Elsayed Ramadan Genena
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
- Department of Clinical Biochemistry, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Maha A F Hamouda
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | - Norhan M Salama
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | - Enas S Zahran
- Rheumatology and Immunology Unit, Internal Medicine Department, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | - Asmaa A Abdel Latif
- Department of Public Health and Community Medicine (Industrial Medicine and Occupational Health Specialty), Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | | |
Collapse
|
28
|
Zhai J, Yuan B, Liu T, Mo L, Xie Y, Zhao Y, Cao S, Meng L. Association between the inflammatory burden index and rheumatoid arthritis and its all-cause mortality: data from NHANES 1999-2018. Front Med (Lausanne) 2024; 11:1421497. [PMID: 39234043 PMCID: PMC11371669 DOI: 10.3389/fmed.2024.1421497] [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: 04/26/2024] [Accepted: 08/08/2024] [Indexed: 09/06/2024] Open
Abstract
Background and aims Rheumatoid arthritis (RA) is a prevalent chronic autoimmune disease characterized by chronic inflammation. The Inflammatory Burden Index (IBI) is a newly proposed comprehensive inflammation index used to assess systemic inflammation. The relationship between IBI and RA, as well as its all-cause mortality, remains unclear. The objective of this study was to examine the correlation between IBI and RA and to analyze the association between IBI and all-cause mortality in RA. Methods The study comprehensively analyzes adult data from the National Health and Nutrition Examination Survey (NHANES) spanning 1999 to 2018. The participants' IBI was calculated using the formula IBI = CRP * neutrophils/lymphocytes. Three models were constructed to investigate the relationship between IBI and the prevalence of RA. Nonlinear relationships were determined using restricted cubic spline curves. Stratified analyses and interaction tests were used to explore the relationship between RA and IBI in different subgroups. The same data analyses were applied to investigate the association between IBI and RA all-cause mortality. Results The data analyses revealed a stable positive and nonlinear correlation between IBI and the risk of RA, as well as a positive, nonlinear, J-shaped association between IBI and RA all-cause mortality. The correlation and association were consistent across most subgroups, and multiple covariates had no effect on the results. No significant effect of multiple covariates on the association was found through interaction tests. Conclusion Our study has demonstrated a positive correlation between the prevalence of RA and all-cause mortality with the IBI index. This suggests that lower levels of inflammation in the body are associated with a reduced risk of RA prevalence and all-cause mortality. Further prospective studies are required to explore the mechanisms involved.
Collapse
Affiliation(s)
- Jiali Zhai
- Department of Biochemistry and Molecular Biology, Institute of Molecular and Translational Medicine (IMTM), Xi'an Jiaotong University Health Science Center, Xi'an, China
- Department of Orthopedics, Civil Aviation General Hospital, Beijing, China
| | - Bo Yuan
- Department of Orthopedics, Civil Aviation General Hospital, Beijing, China
| | - Tiebing Liu
- Civil Aviation Public Health Emergency Management Office, Civil Aviation Medicine Center, Civil Aviation Administration of China, Civil Aviation General Hospital, Beijing, China
| | - Linfei Mo
- Department of Rheumatology and Immunology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yajie Xie
- Department of Biochemistry and Molecular Biology, Institute of Molecular and Translational Medicine (IMTM), Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, and Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shuai Cao
- Department of Orthopedics, Civil Aviation General Hospital, Beijing, China
| | - Liesu Meng
- Department of Biochemistry and Molecular Biology, Institute of Molecular and Translational Medicine (IMTM), Xi'an Jiaotong University Health Science Center, Xi'an, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, China
| |
Collapse
|
29
|
Wu Y, Zhang Y, Wang Z, Lu Y, Wang Y, Pan J, Liu C, Zhu W, Wang Y. Bitongqing Attenuates CIA Rats by Suppressing Macrophage Pyroptosis and Modulating the NLRP3/Caspase-1/GSDMD Pathway. J Inflamm Res 2024; 17:5453-5469. [PMID: 39165322 PMCID: PMC11335010 DOI: 10.2147/jir.s466624] [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: 04/04/2024] [Accepted: 08/02/2024] [Indexed: 08/22/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovitis and inflammatory cell infiltration. The traditional Chinese medicine prescription, Bitongqing (BTQ) exhibited significant efficacy in the clinical treatment of RA. However, the potential therapeutic mechanisms of BTQ in treating RA have not been fully investigated. This study aims to elucidate the effect of BTQ on collagen-induced arthritis (CIA) rat macrophage pyroptosis, providing a theoretical basis for treating RA. Methods This research employed liquid chromatography-mass spectrometry (LC-MS) to identify the primary components of BTQ. The therapeutic effects of BTQ were evaluated in a rat model of CIA. In vivo experiments were conducted using pathohistological staining, immunofluorescence, micro-CT, and Western blotting. Next, Mouse leukemia cells of monocyte macrophage cells (RAW264.7) were induced to undergo pyroptosis using lipopolysaccharide (LPS) and adenosine triphosphate (ATP), and the impact of BTQ on RAW264.7 macrophages was assessed through cell viability, immunofluorescence analysis, lactate dehydrogenase (LDH) secretion measurement, and Western blotting. Results BTQ had a therapeutic effect on CIA rats, which was mainly manifested as a reduction in joint inflammation, foot swelling, bone erosion, and amelioration of pathological changes in these rats. Further studies revealed that BTQ inhibited the levels of cytokine production interleukin-18 (IL-18) and interleukin-1β (IL-1β), and likewise, it inhibited the expression of key proteins in the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) mediated pyroptosis in the synovial tissues of CIA rats. The results of in vitro experiments demonstrated that BTQ attenuated LDH secretion, decreased IL-18 and IL-1β cytokine production, and downregulated expression of key proteins involved in the NLRP3-mediated pyroptosis on RAW264.7 macrophages. Conclusion The therapeutic potential of BTQ in CIA lies in its ability to inhibit NLRP3-mediated macrophage pyroptosis, thereby suggesting a promising strategy for the treatment of RA.
Collapse
Affiliation(s)
- Yunxia Wu
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yue Zhang
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Zishan Wang
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yun Lu
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yabei Wang
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Jie Pan
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Chenxi Liu
- Academy of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Wen Zhu
- Department of Rheumatology & Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yue Wang
- Department of Rheumatology & Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|
30
|
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
|
31
|
Yu Z, Li P, Gao D, Hu Y, Xia F, Liu L, Liu J, Liu W, Zhang H. Inhibition of LSD1 via SP2509 attenuated the progression of rheumatoid arthritis. Immunol Res 2024; 72:797-810. [PMID: 38722530 DOI: 10.1007/s12026-024-09486-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/29/2024] [Indexed: 08/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial hyperplasia, pannus formation, and cartilage and bone destruction. Lysine-specific demethylase 1 (LSD1), an enzyme involved in transcriptional regulation, has an unclear role in synovial inflammation, fibroblast-like synoviocytes migration, and invasion during RA pathogenesis. In this study, we observed increased LSD1 expression in RA synovial tissues and in TNF-α-stimulated MH7A cells. SP2509, an LSD1 antagonist, directly reduced LSD1 expression and reversed the elevated levels of proteins associated with inflammation, apoptosis, proliferation, and autophagy induced by TNF-α. Furthermore, SP2509 inhibited the migratory capacity of MH7A cells, which was enhanced by TNF-α. In CIA models, SP2509 treatment ameliorated RA development, reducing the expression of pro-inflammatory cytokines and alleviating joint pathological symptoms. These findings underscore the significance of LSD1 in RA and propose the therapeutic potential of SP2509.
Collapse
Affiliation(s)
- Ziliang Yu
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China
| | - Peipei Li
- Department of Operating Room, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, 226000, Jiangsu, China
| | - Dagong Gao
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China
| | - Yalong Hu
- Department of Orthopaedics, Qidong People's Hospital, Affiliated Qidong Hospital of Nantong University, Nantong, 226000, Jiangsu, China
| | - Fei Xia
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China
| | - Lei Liu
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China
| | - Jian Liu
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China
| | - Wei Liu
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China.
| | - Haiping Zhang
- Department of Orthopaedics, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Nantong, 226000, Jiangsu, China.
| |
Collapse
|
32
|
Yin X, Zhang Y, Zou J, Yang J. Association of the systemic immune-inflammation index with all-cause and cardiovascular mortality in individuals with rheumatoid arthritis. Sci Rep 2024; 14:15129. [PMID: 38956376 PMCID: PMC11219888 DOI: 10.1038/s41598-024-66152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
The systemic immune-inflammation index (SII), a metric reflecting systemic inflammatory response and immune activation, remains underexplored concerning its correlation with mortality among rheumatoid arthritis (RA) patients. This study aimed to delineate the association between SII and both all-cause and cardiovascular mortality within the cohort of American adults diagnosed with RA, utilizing data from the National Health and Nutrition Examination Survey (NHANES) spanning 1999 to 2018. The investigation extracted data from NHANES cycles between 1999 and 2018, identifying RA patients through questionnaire responses. The SII was computed based on complete blood counts, employing the formula: (platelets × neutrophils) / lymphocytes. The optimal SII cutoff value for significant survival outcomes was determined using maximally selected rank statistics. Multivariable Cox proportional hazards models assessed the relationship between SII levels and mortality (all-cause and cardiovascular) among RA patients, with subgroup analyses examining potential modifications by clinical confounders. Additionally, restricted cubic spline (RCS) analyses were conducted to explore the linearity of the SII-mortality association. The study encompassed 2070 American adults with RA, among whom 287 exhibited a higher SII (≥ 919.75) and 1783 a lower SII (< 919.75). Over a median follow-up duration of 108 months, 602 participants died. After adjustments for demographic, socioeconomic, and lifestyle variables, a higher SII was associated with a 1.48-fold increased risk of all-cause mortality (hazard ratio [HR] = 1.48, 95% confidence interval [CI] 1.21-1.81, P < 0.001) and a 1.51-fold increased risk of cardiovascular mortality (HR = 1.51, 95% CI 1.04-2.18, P = 0.030) compared to a lower SII. Kaplan-Meier analyses corroborated significantly reduced survival rates within the higher SII cohort for both all-cause and cardiovascular mortality (Pall-cause mortality < 0.0001 and Pcardiovascular mortality = 0.0004). RCS analyses confirmed a positive nonlinear relationship between SII and mortality rates. In conclusion, the SII offers a straightforward indicator of the equilibrium between detrimental innate inflammation and beneficial adaptive immunity. Our investigation, utilizing a comprehensive and nationally representative sample, reveals that elevated SII levels independently forecast a greater risk of mortality from all causes, as well as cardiovascular-specific mortality, in individuals suffering from RA. These insights underscore the clinical relevance of the SII as an affordable and readily accessible biomarker. Its incorporation into regular clinical practice could significantly enhance the precision of risk assessment and forecasting for patients with RA, facilitating more tailored and effective management strategies. Specifically, patients with high SII levels could be identified for more stringent cardiovascular risk management, including closer monitoring, lifestyle interventions, and aggressive pharmacological treatments to mitigate their increased risk of mortality.
Collapse
Affiliation(s)
- Xiaoshuang Yin
- Department of Immunology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Yu Zhang
- Department of Immunology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China
| | - Jinmei Zou
- Department of Immunology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China.
| | - Jing Yang
- Department of Immunology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, China.
| |
Collapse
|
33
|
Li C, Sun Y, Xu W, Chang F, Wang Y, Ding J. Mesenchymal Stem Cells-Involved Strategies for Rheumatoid Arthritis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305116. [PMID: 38477559 PMCID: PMC11200100 DOI: 10.1002/advs.202305116] [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: 07/26/2023] [Revised: 12/13/2023] [Indexed: 03/14/2024]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic inflammation of the joints and bone destruction. Because of systemic administration and poor targeting, traditional anti-rheumatic drugs have unsatisfactory treatment efficacy and strong side effects, including myelosuppression, liver or kidney function damage, and malignant tumors. Consequently, mesenchymal stem cells (MSCs)-involved therapy is proposed for RA therapy as a benefit of their immunosuppressive and tissue-repairing effects. This review summarizes the progress of MSCs-involved RA therapy through suppressing inflammation and promoting tissue regeneration and predicts their potential clinical application.
Collapse
Affiliation(s)
- Chaoyang Li
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Yifu Sun
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Weiguo Xu
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Fei Chang
- Department of OrthopedicsThe Second Hospital of Jilin University4026 Yatai StreetChangchun130041P. R. China
| | - Yinan Wang
- Department of BiobankDivision of Clinical ResearchThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of EducationThe First Hospital of Jilin University1 Xinmin StreetChangchun130061P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
34
|
Parab A, Bhatt LK. T-cell metabolism in rheumatoid arthritis: focus on mitochondrial and lysosomal dysfunction. Immunopharmacol Immunotoxicol 2024; 46:378-384. [PMID: 38478010 DOI: 10.1080/08923973.2024.2330645] [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: 06/07/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by immune cell dysregulation, synovial hyperplasia, and progressive cartilage destruction. The loss of immunological self-tolerance against autoantigens is the crucial insult responsible for the pathogenesis of RA. These immune abnormalities are experienced many years before the onset of clinical arthritis. OBJECTIVE This review aims to discuss the metabolic status of T-cells in RA and focuses mainly on mitochondrial and lysosomal dysfunctions involved in altering the T-cell metabolism. DISCUSSION T-cells are identified as the primary initiators of immunological abnormalities in RA. These RA T-cells show a distinct metabolic pattern compared to the healthy individuals. Dampened glycolytic flux, poor ATP production, and shifting of glucose to the pentose phosphate pathway resulting in increased NADPH and decreased ROS levels are the common metabolic patterns observed in RA T-cells. Defective mtDNA due to lack of MRE11A gene, a key molecular actor for resection, and inefficient lysosomal function due to misplacement of AMPK on the lysosomal surface were found to be responsible for mitochondrial and lysosome dysfunction in RA. Targeting this mechanism in RA can alleviate aggressive T-cell phenotype and may control the severity of RA.
Collapse
Affiliation(s)
- Asmita Parab
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
35
|
Huang Z, Chen C, Tan L, Ling Y, Ma W, Zhang J. 16S rRNA Gene Sequencing of Gut Microbiota in Rheumatoid Arthritis Treated with 99Tc-MDP. Pharmgenomics Pers Med 2024; 17:237-249. [PMID: 38807628 PMCID: PMC11130713 DOI: 10.2147/pgpm.s451065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/27/2024] [Indexed: 05/30/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a common autoimmune disease with the main symptoms being joint swelling and pain. In severe cases, joint deformity or even complete loss of function occurs. Technetium methylene diphosphonate (99Tc-MDP) is widely used for RA treatment in China, but there are no studies on the effects of 99Tc-MDP on intestinal flora. Objective To explore the effects of 99Tc-MDP treatment on the composition and function of the intestinal flora and to provide new information on the mechanism of 99Tc-MDP in RA treatment. Methods Stool samples from RA patients before and after 99Tc-MDP treatment were collected to form two groups (Before and After). Total genomic DNA of the samples was extracted for 16S rRNA gene sequencing. The altered composition of the intestinal flora, the key target bacteria regulated by 99Tc-MDP, and the pathways of action of 99 Tc-MDP were analyzed by bioinformatics. Results A total of 64 fresh stool samples were collected from 32 RA patients. Compared to the Before group, the After group showed increased Bacteroidetes abundance and decreased Firmicutes abundance. At the genus level, Prevotella increased whereas Escherichia decreased. Both α and β diversity analyses showed that 99Tc-MDP treatment did not affect gut microbial diversity in RA patients. LEfSe analyses and random forest analyses showed Bacteroidetes, Prevotella, Enterococcus, Escherichia and Ruminococcaceae were the main 99Tc-MDP regulating bacteria. Functional enrichment analysis revealed that the functional differences in gut flora of the two groups centered on Metabolism and Genetic Information Processing. Conclusion This study revealed differences in the composition of the gut microbiota in RA patients before and after 99Tc-MDP treatment. The therapeutic effect of 99Tc MDP is mainly achieved through Bacteroidetes, Prevotella, and Enterococcus. Regulating metabolism and genetic information processing of gut flora may be the mechanism of 99Tc-MDP in treating RA.
Collapse
Affiliation(s)
- Zhaowei Huang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
| | - Changming Chen
- Department of Rheumatology & Immunology, The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, Guizhou, People’s Republic of China
| | - Linlin Tan
- Department of Internal Medicine, Jinshazhou Hospital Affiliated to Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People’s Republic of China
| | - Yi Ling
- Department of Rheumatology & Immunology, The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, Guizhou, People’s Republic of China
| | - Wukai Ma
- Department of Rheumatology & Immunology, The Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang, Guizhou, People’s Republic of China
| | - Jinan Zhang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Department of Endocrinology & Rheumatology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
36
|
Yang J, Wei Z, Li H, Lv S, Fu Y, Xiao L. Paeoniflorin inhibits the inflammation of rheumatoid arthritis fibroblast-like synoviocytes by downregulating hsa_circ_009012. Heliyon 2024; 10:e30555. [PMID: 38726183 PMCID: PMC11079323 DOI: 10.1016/j.heliyon.2024.e30555] [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: 01/27/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to progressive joint damage. Circular RNA (circRNA) can regulate the inflammatory response of fibroblast-like synoviocytes (FLSs) in RA, influencing the disease progression. Paeoniflorin (PF) is the main active ingredient extracted from Paeonia lactiflora and is known for its anti-inflammatory effect. This study aims to explore the potential mechanisms by which hsa_circ_009012 and PF regulate the inflammatory response in RA. Methods RNA expression of hsa_circ_009012, has-microRNA-1286 (miR-1286), toll-like receptor 4 (TLR4), NOD-like receptor thermal protein domain associated protein 3 (NLRP3) was assessed by real-time quantitative polymerase chain reaction (RT-qPCR) or western blotting (WB). Cell inflammation markers (TNF-α, IL-1β, IL-6) were assessed by RT-qPCR and immunofluorescence (IF). Counting Kit-8 (CCK-8) assay, flow cytometry, and transwell assay were utilized to test cell viability, cell cycle distribution, and migration. Results Hsa_circ_009012 was highly expressed in RA-FLS. Hsa_circ_009012 over-expression facilitated the inflammation in RA-FLS and was closely associated with the miR-1286/TLR4 axis. Paeoniflorin inhibited inflammation and the expression of hsa_circ_009012 and TLR4, while upregulating the expression of miR-1286 in RA-FLS. Moreover, the upregulation of hsa_circ_009012 reversed the repressive effect of paeoniflorin on RA-FLS progression. Conclusion Paeoniflorin inhibits the inflammation of RA-FLS via mediating the hsa_circ_009012/miR-1286/TLR4/NLRP3 axis.
Collapse
Affiliation(s)
- Junping Yang
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
- Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Zehong Wei
- Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Huaiyu Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
- Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Senhao Lv
- Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yong Fu
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
- Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Liang Xiao
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, 330006, Jiangxi, People's Republic of China
| |
Collapse
|
37
|
More NE, Mandlik R, Zine S, Gawali VS, Godad AP. Exploring the therapeutic opportunities of potassium channels for the treatment of rheumatoid arthritis. Front Pharmacol 2024; 15:1286069. [PMID: 38783950 PMCID: PMC11111972 DOI: 10.3389/fphar.2024.1286069] [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: 08/30/2023] [Accepted: 01/18/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the synovial joint, which leads to inflammation, loss of function, joint destruction, and disability. The disease biology of RA involves complex interactions between genetic and environmental factors and is strongly associated with various immune cells, and each of the cell types contributes differently to disease pathogenesis. Several immunomodulatory molecules, such as cytokines, are secreted from the immune cells and intervene in the pathogenesis of RA. In immune cells, membrane proteins such as ion channels and transporters mediate the transport of charged ions to regulate intracellular signaling pathways. Ion channels control the membrane potential and effector functions such as cytotoxic activity. Moreover, clinical studies investigating patients with mutations and alterations in ion channels and transporters revealed their importance in effective immune responses. Recent studies have shown that voltage-gated potassium channels and calcium-activated potassium channels and their subtypes are involved in the regulation of immune cells and RA. Due to the role of these channels in the pathogenesis of RA and from multiple pieces of clinical evidence, they can be considered therapeutic targets for the treatment of RA. Here, we describe the role of voltage-gated and calcium-activated potassium channels and their subtypes in RA and their pharmacological application as drug targets.
Collapse
Affiliation(s)
| | - Rahul Mandlik
- Medical Affairs, Shalina Healthcare DMCC, Dubai, United Arab Emirates
| | - Sandip Zine
- SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | | | - Angel Pavalu Godad
- SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
38
|
Malik S, Chakraborty D, Agnihotri P, Sharma A, Biswas S. Mitochondrial functioning in Rheumatoid arthritis modulated by estrogen: Evidence-based insight into the sex-based influence on mitochondria and disease. Mitochondrion 2024; 76:101854. [PMID: 38403096 DOI: 10.1016/j.mito.2024.101854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Alteration of immune response and synovium microvasculature in Rheumatoid arthritis (RA) progression has been suggested to be associated with mitochondrial functioning. Mitochondria, with maternally inherited DNA, exhibit differential response to the female hormone estrogen. Various epidemiological evidence has also shown the prominence of RA in the female population, depicting the role of estrogen in modulating the pathogenesis of RA. As estrogen regulates the expression of differential proteins and associated signaling pathways of RA, its influence on mitochondrial functioning seems evident. Thus, in this review, the studies related to mitochondria and their relation with estrogen and Rheumatoid arthritis were retrieved. We analyzed the different mitochondrial activities that are altered in RA and the possibility of their estrogenic control. The study expands to in silico analysis, revealing the differential mitochondrial proteins expressed in RA and examining these proteins as potential estrogenic targets. It was found that ALDH2, CASP3, and SOD2 are the major mitochondrial proteins involved in RA progression and are also potent estradiol targets. The analysis establishes the role of mitochondrial proteins in RA progression, which were found to be direct or indirect targets of estrogen, depicting its potential for regulating mitochondrial functions in RA.
Collapse
Affiliation(s)
- Swati Malik
- Department of Integrative and Functional Biology, CSIR - Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; AcSIR - Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Debolina Chakraborty
- Department of Integrative and Functional Biology, CSIR - Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; AcSIR - Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Prachi Agnihotri
- Department of Integrative and Functional Biology, CSIR - Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; AcSIR - Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Alankrita Sharma
- Department of Integrative and Functional Biology, CSIR - Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | - Sagarika Biswas
- Department of Integrative and Functional Biology, CSIR - Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India; AcSIR - Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
39
|
Wan W, Qian X, Zhou B, Gao J, Deng J, Zhao D. Integrative analysis and validation of necroptosis-related molecular signature for evaluating diagnosis and immune features in Rheumatoid arthritis. Int Immunopharmacol 2024; 131:111809. [PMID: 38484666 DOI: 10.1016/j.intimp.2024.111809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease that is characterized by persistent morning stiffness, joint pain, and swelling. However, there is a lack of reliable diagnostic markers and therapeutic targets that are both effective and trustworthy. METHODS In this study, gene expression profiles (GSE89408, GSE55235, GSE55457, and GSE77298) were obtained from the Gene Expression Omnibus database. Differentially expressed necroptosis-related genes were attained from intersection of necroptosis-related gene set, differentially expressed genes, and weighted gene co-expression network analysis. The LASSO, random forest, and SVM-RFE machine learning algorithms were utilized to further screen potential diagnostic genes for RA. Immune cell infiltration was analyzed using the CIBERSORT method. The expressions of diagnostic genes were validated through quantitative real-time PCR, western blotting, immunohistochemistry, and immunofluorescence staining in synovial tissues collected from three trauma controls and three RA patients. RESULTS Five core necroptosis-related genes (FAS, CYBB, TNFSF10, EIF2AK2, and BIRC2) were identified as potential biomarkers for RA. Two different necroptosis patterns based on these five genes were confirmed to significantly correlated with immune cells (especially macrophages). In vitro experiments showed significantly higher mRNA and protein expression levels of CYBB and EIF2AK2 in RA patients compared to normal controls, consistent with the bioinformatics analysis results. CONCLUSION Our study identified a novel necroptosis-related subtype and five diagnostic biomarkers of RA, revealed vital roles in the development and occurrence of RA, and offered potential targets for clinical diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Wei Wan
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Xinyu Qian
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Bole Zhou
- Department of Joint Bone Disease Surgery, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Jie Gao
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China
| | - Jiewen Deng
- Department of Cardiovascular Diseases, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China.
| | - Dongbao Zhao
- Department of Rheumatology and Immunology, Shanghai Changhai Hospital, the first affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
40
|
Pant T, Uche N, Juric M, Zielonka J, Bai X. Regulation of immunomodulatory networks by Nrf2-activation in immune cells: Redox control and therapeutic potential in inflammatory diseases. Redox Biol 2024; 70:103077. [PMID: 38359749 PMCID: PMC10877431 DOI: 10.1016/j.redox.2024.103077] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
Inflammatory diseases present a serious health challenge due to their widespread prevalence and the severe impact on patients' lives. In the quest to alleviate the burden of these diseases, nuclear factor erythroid 2-related factor 2 (Nrf2) has emerged as a pivotal player. As a transcription factor intimately involved in cellular defense against metabolic and oxidative stress, Nrf2's role in modulating the inflammatory responses of immune cells has garnered significant attention. Recent findings suggest that Nrf2's ability to alter the redox status of cells underlies its regulatory effects on immune responses. Our review delves into preclinical and clinical evidence that underscores the complex influence of Nrf2 activators on immune cell phenotypes, particularly in the inflammatory milieu. By offering a detailed analysis of Nrf2's role in different immune cell populations, we cast light on the potential of Nrf2 activators in shaping the immune response towards a more regulated state, mitigating the adverse effects of inflammation through modeling redox status of immune cells. Furthermore, we explore the innovative use of nanoencapsulation techniques that enhance the delivery and efficacy of Nrf2 activators, potentially advancing the treatment strategies for inflammatory ailments. We hope this review will stimulate the development and expansion of Nrf2-targeted treatments that could substantially improve outcomes for patients suffering from a broad range of inflammatory diseases.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Nnamdi Uche
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
41
|
Paramasivam S, Perumal SS, Ekambaram SP. Computational Deciphering of the Role of S100A8 and S100A9 Proteins and Their Changes in the Structure Assembly Influences Their Interaction with TLR4, RAGE, and CD36. Protein J 2024; 43:243-258. [PMID: 38431537 DOI: 10.1007/s10930-024-10186-0] [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] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
S100A8 and S100A9 belong to the calcium-binding, damage associated molecular pattern (DAMP) proteins shown to aggravate the pathogenesis of rheumatoid arthritis (RA) through their interaction with the TLR4, RAGE and CD36 receptors. S100A8 and S100A9 proteins tend to exist in monomeric, homo and heterodimeric forms, which have been implicated in the pathogenesis of RA, via interacting with Pattern Recognition receptors (PRRs). The study aims to assess the influence of changes in the structure and biological assembly of S100A8 and S100A9 proteins as well as their interaction with significant receptors in RA through computational methods and surface plasmon resonance (SPR) analysis. Molecular docking analysis revealed that the S100A9 homodimer and S100A8/A9 heterodimer showed higher binding affinity towards the target receptors. Most S100 proteins showed good binding affinity towards TLR4 compared to other receptors. Based on the 50 ns MD simulations, TLR4, RAGE, and CD36 formed stable complexes with the monomeric and dimeric forms of S100A8 and S100A9 proteins. However, SPR analysis showed that the S100A8/A9 heterodimers formed stable complexes and exhibited high binding affinity towards the receptors. SPR data also indicated that TLR4 and its interactions with S100A8/A9 proteins may play a primary role in the pathogenesis of RA, with additional contributions from CD36 and RAGE interactions. Subsequent in vitro and in vivo investigations are warranted to corroborate the involvement of S100A8/A9 and the expression of TLR4, RAGE, and CD36 in the pathophysiology of RA.
Collapse
Affiliation(s)
- Sivasakthi Paramasivam
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Senthamil Selvan Perumal
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India
| | - Sanmuga Priya Ekambaram
- Department of Pharmaceutical Technology, Bharathidasan Institute of Technology Campus, University College of Engineering, Anna University, Tiruchirappalli, Tamil Nadu, 620 024, India.
| |
Collapse
|
42
|
Ugolkov Y, Nikitich A, Leon C, Helmlinger G, Peskov K, Sokolov V, Volkova A. Mathematical modeling in autoimmune diseases: from theory to clinical application. Front Immunol 2024; 15:1371620. [PMID: 38550585 PMCID: PMC10973044 DOI: 10.3389/fimmu.2024.1371620] [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: 01/16/2024] [Accepted: 02/29/2024] [Indexed: 04/02/2024] Open
Abstract
The research & development (R&D) of novel therapeutic agents for the treatment of autoimmune diseases is challenged by highly complex pathogenesis and multiple etiologies of these conditions. The number of targeted therapies available on the market is limited, whereas the prevalence of autoimmune conditions in the global population continues to rise. Mathematical modeling of biological systems is an essential tool which may be applied in support of decision-making across R&D drug programs to improve the probability of success in the development of novel medicines. Over the past decades, multiple models of autoimmune diseases have been developed. Models differ in the spectra of quantitative data used in their development and mathematical methods, as well as in the level of "mechanistic granularity" chosen to describe the underlying biology. Yet, all models strive towards the same goal: to quantitatively describe various aspects of the immune response. The aim of this review was to conduct a systematic review and analysis of mathematical models of autoimmune diseases focused on the mechanistic description of the immune system, to consolidate existing quantitative knowledge on autoimmune processes, and to outline potential directions of interest for future model-based analyses. Following a systematic literature review, 38 models describing the onset, progression, and/or the effect of treatment in 13 systemic and organ-specific autoimmune conditions were identified, most models developed for inflammatory bowel disease, multiple sclerosis, and lupus (5 models each). ≥70% of the models were developed as nonlinear systems of ordinary differential equations, others - as partial differential equations, integro-differential equations, Boolean networks, or probabilistic models. Despite covering a relatively wide range of diseases, most models described the same components of the immune system, such as T-cell response, cytokine influence, or the involvement of macrophages in autoimmune processes. All models were thoroughly analyzed with an emphasis on assumptions, limitations, and their potential applications in the development of novel medicines.
Collapse
Affiliation(s)
- Yaroslav Ugolkov
- Research Center of Model-Informed Drug Development, Ivan Mikhaylovich (I.M.) Sechenov First Moscow State Medical University, Moscow, Russia
- Marchuk Institute of Numerical Mathematics of the Russian Academy of Sciences (RAS), Moscow, Russia
| | - Antonina Nikitich
- Research Center of Model-Informed Drug Development, Ivan Mikhaylovich (I.M.) Sechenov First Moscow State Medical University, Moscow, Russia
- Marchuk Institute of Numerical Mathematics of the Russian Academy of Sciences (RAS), Moscow, Russia
| | - Cristina Leon
- Modeling and Simulation Decisions FZ - LLC, Dubai, United Arab Emirates
| | | | - Kirill Peskov
- Research Center of Model-Informed Drug Development, Ivan Mikhaylovich (I.M.) Sechenov First Moscow State Medical University, Moscow, Russia
- Marchuk Institute of Numerical Mathematics of the Russian Academy of Sciences (RAS), Moscow, Russia
- Modeling and Simulation Decisions FZ - LLC, Dubai, United Arab Emirates
- Sirius University of Science and Technology, Sirius, Russia
| | - Victor Sokolov
- Marchuk Institute of Numerical Mathematics of the Russian Academy of Sciences (RAS), Moscow, Russia
- Modeling and Simulation Decisions FZ - LLC, Dubai, United Arab Emirates
| | - Alina Volkova
- Marchuk Institute of Numerical Mathematics of the Russian Academy of Sciences (RAS), Moscow, Russia
- Modeling and Simulation Decisions FZ - LLC, Dubai, United Arab Emirates
| |
Collapse
|
43
|
Rufino AT, Freitas M, Proença C, Ferreira de Oliveira JMP, Fernandes E, Ribeiro D. Rheumatoid arthritis molecular targets and their importance to flavonoid-based therapy. Med Res Rev 2024; 44:497-538. [PMID: 37602483 DOI: 10.1002/med.21990] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/18/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
Rheumatoid arthritis (RA) is a progressive, chronic, autoimmune, inflammatory, and systemic condition that primarily affects the synovial joints and adjacent tissues, including bone, muscle, and tendons. The World Health Organization recognizes RA as one of the most prevalent chronic inflammatory diseases. In the last decade, there was an expansion on the available RA therapeutic options which aimed to improve patient's quality of life. Despite the extensive research and the emergence of new therapeutic approaches and drugs, there are still significant unwanted side effects associated to these drugs and still a vast number of patients that do not respond positively to the existing therapeutic strategies. Over the years, several references to the use of flavonoids in the quest for new treatments for RA have emerged. This review aimed to summarize the existing literature about the flavonoids' effects on the major pathogenic/molecular targets of RA and their potential use as lead compounds for the development of new effective molecules for RA treatment. It is demonstrated that flavonoids can modulate various players in synovial inflammation, regulate immune cell function, decrease synoviocytes proliferation and balance the apoptotic process, decrease angiogenesis, and stop/prevent bone and cartilage degradation, which are all dominant features of RA. Although further investigation is necessary to determine the effectiveness of flavonoids in humans, the available data from in vitro and in vivo models suggest their potential as new disease-modifying anti-rheumatic drugs. This review highlights the use of flavonoids as a promising avenue for future research in the treatment of RA.
Collapse
Affiliation(s)
- Ana T Rufino
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Carina Proença
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - José M P Ferreira de Oliveira
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Daniela Ribeiro
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
- Faculty of Agrarian Sciences and Environment, University of the Azores, Açores, Portugal
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Type 1 interferons (IFN-I) are of increasing interest across a wide range of autoimmune rheumatic diseases. Historically, research into their role in rheumatoid arthritis (RA) has been relatively neglected, but recent work continues to highlight a potential contribution to RA pathophysiology. RECENT FINDINGS We emphasise the importance of disease stage when examining IFN-I in RA and provide an overview on how IFN-I may have a direct role on a variety of relevant cellular functions. We explore how clinical trajectory may be influenced by increased IFN-I signalling, and also, the limitations of scores composed of interferon response genes. Relevant environmental triggers and inheritable RA genetic risk relating to IFN-I signalling are explored with emphasis on intriguing data potentially linking IFN-I exposure, epigenetic changes, and disease relevant processes. Whilst these data cumulatively illustrate a likely role for IFN-I in RA, they also highlight the knowledge gaps, particularly in populations at risk for RA, and suggest directions for future research to both better understand IFN-I biology and inform targeted therapeutic strategies.
Collapse
Affiliation(s)
- Chung M A Lin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Faye A H Cooles
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
- Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
45
|
Madahar SS, Gideon A, Abdul-Sater AA. Nod-like receptors in inflammatory arthritis. Biomed J 2024; 47:100655. [PMID: 37598797 PMCID: PMC10825342 DOI: 10.1016/j.bj.2023.100655] [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: 06/22/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/22/2023] Open
Abstract
Nod-like receptors (NLRs) are innate immune receptors that play a key role in sensing components from pathogens and from damaged cells or organelles. NLRs form signaling complexes that can lead to activation of transcription factors or effector caspases - by means of inflammasome activation -Inflammatory arthritis (IA) culminating in promoting inflammation. An increasing body of research supports the role of NLRs in driving pathogenesis of IA, a collection of diseases that include rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis, and pediatric arthritis. In this review, we briefly discuss the main drivers of IA diseases and dive into the evidence for - and against - various NLRs in driving these diseases. We also review the studies examining the use of NLR and inflammasome inhibitors as potential therapies for IA.
Collapse
Affiliation(s)
- Sahib Singh Madahar
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada; Department of Biology, York University, Toronto, Ontario, Canada
| | - Alita Gideon
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada
| | - Ali A Abdul-Sater
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario, Canada.
| |
Collapse
|
46
|
Kattner AA. Evolutionary edge: NOD-like receptors in immunity. Biomed J 2024; 47:100702. [PMID: 38301953 PMCID: PMC10885312 DOI: 10.1016/j.bj.2024.100702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024] Open
Abstract
This issue of the Biomedical Journal delves into the multifaceted roles of NOD-like receptors (NLRs) in immunity, examining their subfamilies and functions within innate and adaptive immunity, autoimmune and inflammatory conditions, and mitophagy regulation. In this issue the dynamics of mRNA vaccines are explored, as well as the synergistic effects of a ketogenic diet with anti-tumor therapies, the roles of curcumin and RANKL in osteoclastogenesis, and the validation of a rapid diagnostic test for an oral cancer biomarker. Additionally, advancements in ocular care are highlighted, featuring a novel prodrug targeting corneal neovascularization, and discussing the efficacy of dexamethasone implants against macular edema. Concluding, further insights into the impact of sweetened foods on child development are given.
Collapse
|
47
|
Gao S, Song H. Integrated comparison of the mRNAome in cartilage, synovium, and macrophages in osteoarthritis. Z Rheumatol 2024; 83:62-70. [PMID: 35178608 DOI: 10.1007/s00393-022-01171-y] [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: 01/21/2022] [Indexed: 11/09/2022]
Abstract
The precise molecular mechanisms associated with osteoarthritis (OA), the most common musculoskeletal disorder, are poorly understood. There are currently no effective treatments to prevent the initiation and progression of the disease. In recent years, the development of mRNAome has made it possible to identify new mechanisms and therapeutic targets. However, the differentially expressed genes screened by different microarrays are not completely the same. In order to avoid this shortcoming, we integrate the different genes from different tissues and data sets, and select the commonly expressed genes for further studies.
Collapse
Affiliation(s)
- Siming Gao
- Department of Rheumatology, Beijing Jishuitan Hospital, No. 31, Xin Jie Kou East Street, Xicheng District, 100035, Beijing, China
| | - Hui Song
- Department of Rheumatology, Beijing Jishuitan Hospital, No. 31, Xin Jie Kou East Street, Xicheng District, 100035, Beijing, China.
| |
Collapse
|
48
|
Xiao T, Cheng X, Zhi Y, Tian F, Wu A, Huang F, Tao L, Guo Z, Shen X. Ameliorative effect of Alangium chinense (Lour.) Harms on rheumatoid arthritis by reducing autophagy with targeting regulate JAK3-STAT3 and COX-2 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117133. [PMID: 37690476 DOI: 10.1016/j.jep.2023.117133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alangium chinense has been used as traditional folk medicine for centuries to treat rheumatoid arthritis (RA) by Guizhou Miao nationality with remarkable clinical effect. But the mechanism of its anti-RA is not fully clarified. AIM OF THE STUDY To explore the effect and underlying mechanism of A. chinense against RA. MATERIAL AND METHODS RA rats were induced by CII/IFA, and oral administrated with or without ethyl acetate extracts of Alangium chinense (ACEE) and tripterygium glycosides (GTW). Then arthritis scores, inflammatory factors in serum and histological evaluation were evaluated to assess the degree of joints disease. Proteomics were conducted via LC-MS/MS to clarify the mechanism of ACEE preliminarily, and further examined by immunohistochemistry, immunofluorescence, western botting, and molecular docking. RESULTS ACEE decreased joints swelling, cell abscission and necrosis of joint tissues arthropathy of RA rats, and attenuated expression of TNF-α, IL-1β, IL-6, PGE2, TGF-β. Meanwhile, differentially expressed proteins in the ACEE treated groups were observed, which were involved in RA, spliceosome, cell adhesion molecules, phagosome and lysosome signaling pathways. Moreover, ACEE significantly ameliorated arthropathy, suppressed JAK-STAT pathway (JAK3, p-JAK3, STAT3, iNOS, RANKL), COX-2 pathway (COX-2, TNF-α, IL-6I, L-1β, 5-LOX), and autophagic signaling pathway (LC3-Ⅰ, LC3-Ⅱ, p62, mTOR). But it showed little effect on the expression of COX-1, JAK1, JAK2, TyK2. CONCLUSION It is the first evidence that A. chinense significantly ameliorates RA, and the underlying immune mechanism involves reducing autophagy with targeting regulate JAK3-STAT3 and COX-2 pathways.
Collapse
Affiliation(s)
- Ting Xiao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Xingyan Cheng
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Yuan Zhi
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Fangfang Tian
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Ai Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Feilong Huang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| | - Zhenghong Guo
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550025, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China; The Key Laboratory of Optimal Utilization of Natural Medicine Resources (The Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550031, China.
| |
Collapse
|
49
|
Zerrouk N, Alcraft R, Hall BA, Augé F, Niarakis A. Large-scale computational modelling of the M1 and M2 synovial macrophages in rheumatoid arthritis. NPJ Syst Biol Appl 2024; 10:10. [PMID: 38272919 PMCID: PMC10811231 DOI: 10.1038/s41540-024-00337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Macrophages play an essential role in rheumatoid arthritis. Depending on their phenotype (M1 or M2), they can play a role in the initiation or resolution of inflammation. The M1/M2 ratio in rheumatoid arthritis is higher than in healthy controls. Despite this, no treatment targeting specifically macrophages is currently used in clinics. Thus, devising strategies to selectively deplete proinflammatory macrophages and promote anti-inflammatory macrophages could be a promising therapeutic approach. State-of-the-art molecular interaction maps of M1 and M2 macrophages in rheumatoid arthritis are available and represent a dense source of knowledge; however, these maps remain limited by their static nature. Discrete dynamic modelling can be employed to study the emergent behaviours of these systems. Nevertheless, handling such large-scale models is challenging. Due to their massive size, it is computationally demanding to identify biologically relevant states in a cell- and disease-specific context. In this work, we developed an efficient computational framework that converts molecular interaction maps into Boolean models using the CaSQ tool. Next, we used a newly developed version of the BMA tool deployed to a high-performance computing cluster to identify the models' steady states. The identified attractors are then validated using gene expression data sets and prior knowledge. We successfully applied our framework to generate and calibrate the M1 and M2 macrophage Boolean models for rheumatoid arthritis. Using KO simulations, we identified NFkB, JAK1/JAK2, and ERK1/Notch1 as potential targets that could selectively suppress proinflammatory macrophages and GSK3B as a promising target that could promote anti-inflammatory macrophages in rheumatoid arthritis.
Collapse
Affiliation(s)
- Naouel Zerrouk
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Rachel Alcraft
- Advanced Research Computing Centre, University College London, London, UK
| | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Franck Augé
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Anna Niarakis
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France.
- Lifeware Group, Inria Saclay, Palaiseau, France.
| |
Collapse
|
50
|
Nakamura Y, Ishibashi HK, Saruga T, Imaizumi T, Kurose A, Tachizaki M, Kawaguchi S, Seya K, Sasaki E, Ishibashi Y. Possible involvement of DExD/H box helicase 60 in synovial inflammation of rheumatoid arthritis: role of toll-like receptor 3 signaling. Mol Biol Rep 2024; 51:131. [PMID: 38236450 DOI: 10.1007/s11033-023-09063-3] [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: 11/17/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Innate immunity is known to be implicated in the etiology of synovitis in rheumatoid arthritis (RA). However, details of the molecular mechanisms have not been fully clarified. DExD/H-box helicase 60 (DDX60), a putative RNA helicase, is of consequence in anti-viral innate immune reactions followed by inflammation. Although DDX60 is involved in the pathogenesis of autoimmune diseases such as systemic lupus nephritis, the role of DDX60 in RA has not been elucidated. The objective of this study was to examine the expression and the role of DDX60 in RA synovial inflammation. METHODS AND RESULTS DDX60 protein expression was investigated by immunohistochemistry in synovial tissues resected from 4 RA and 4 osteoarthritis (OA) patients. We found that synovial DDX60 expression was more intense in RA than in OA. Treatment of human rheumatoid fibroblast-like synoviocytes in culture with polyinosinic-polycytidylic acid, a Toll-like receptor 3 (TLR3) ligand, increased DDX60 protein and mRNA expression. A knockdown experiment of DDX60 using RNA interference revealed a decrease in the expression of poly IC-induced C-X-C motif chemokine ligand 10 (CXCL10) which induces lymphocyte chemotaxis. CONCLUSIONS The synovial DDX60 was more expressed in RA patients than in OA. In human RFLS, DDX60 stimulated by TLR3 signaling affected CXCL10 expression. DDX60 may contribute to synovial inflammation in RA.
Collapse
Affiliation(s)
- Yuzuru Nakamura
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan.
| | - Hikaru Kristi Ishibashi
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Tatsuro Saruga
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Tadaatsu Imaizumi
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Akira Kurose
- Department of Anatomic Pathology, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Mayuki Tachizaki
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Shogo Kawaguchi
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Kazuhiko Seya
- Department of Vascular Biology, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Eiji Sasaki
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| | - Yasuyuki Ishibashi
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, 036-8562, Japan
| |
Collapse
|