1
|
Wu G, Su T, Zhou P, Tang R, Zhu X, Wang J, Chao M, Fan L, Yan H, Ye P, Yu D, Gao F, Chen H. Engineering M2 macrophage-derived exosomes modulate activated T cell cuproptosis to promote immune tolerance in rheumatoid arthritis. Biomaterials 2025; 315:122943. [PMID: 39509857 DOI: 10.1016/j.biomaterials.2024.122943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Nanomedicines for immune modulation have made advancements in the treatment of rheumatoid arthritis (RA). However, due to aberrations in patients' immune systems, inducing antigen-specific immune tolerance while halting disease progression remains a significant challenge. Here, we develop a highly targeted multifunctional nanocomplex, termed M2Exo@CuS-CitP-Rapa (M2CPR), with the aim of selectively inhibiting inflammatory immune reactions while promoting immune tolerance towards specific antigens. M2CPR specifically targets inflammatory tissues in RA, delivering CuS NPs, CitP, Rapa, and endogenous anti-inflammatory factors, thereby ameliorating the inflammatory joint microenvironment. CuS NPs induce Cuproptosis of activated T cells, whose fragments are engulfed by resident or recruited macrophages, resulting in abundant production of TGF-β. TGF-β acts synergistically with Rapa to induce the iDCs into tDCs. tDCs present CitP to Naive T cells, promoting Tregs differentiation. Tregs, in turn, produce more TGF-β, inducing tDCs differentiation, thereby establishing a cycle of immune tolerance. Through in vitro and in vivo experiments, we validate that M2CPR can induce robust and durable antigen-specific immune tolerance, offering a new paradigm for RA therapy.
Collapse
Affiliation(s)
- Guoquan Wu
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Tianyu Su
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peng Zhou
- Department of Orthopedics, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Rongze Tang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xu Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jin Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Minghao Chao
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Liying Fan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Hanrong Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peng Ye
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Dehong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Hongliang Chen
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
2
|
Gao A, Wu R, Mu Y, Jin R, Jiang S, Gao C, Li X, Wang C. Restoring immune tolerance in pre-RA: immunometabolic dialogue between gut microbiota and regulatory T cells. Front Immunol 2025; 16:1565133. [PMID: 40181974 PMCID: PMC11965651 DOI: 10.3389/fimmu.2025.1565133] [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: 01/22/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Rheumatoid arthritis (RA) is a complex chronic autoimmune disease that remains incurable for most patients. With advances in our understanding of the disease's natural history, the concept of pre-RA has emerged as a window of opportunity to intervene before irreversible joint damage occurs. Numerous studies have indicated that the key step driving autoimmunity in early pre-RA lies at an extra-articular site, which is closely related to the regulatory T (Treg) cell-established immune tolerance to the gut microbiota. The intricate immunometabolic crosstalk between Treg cells and the gut microbiota is beginning to be understood, with the re-recognition of Treg cells as metabolic sensors in recent years. In the future, deciphering their immunometabolic dialogue may help to elucidate the underlying mechanisms of pre-RA. Identifying novel biological pathways in the pre-RA stage will bring insights into restoring immune tolerance, thereby potentially curing or preventing the onset of RA.
Collapse
Affiliation(s)
- Anqi Gao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Yanfei Mu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Ruqing Jin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Saixin Jiang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Chong Gao
- Pathology, Joint Program in Transfusion Medicine, Brigham and Women’s Hospital/Children’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Department of Rheumatology, Shanxi Precision Medical Engineering Research Center for Rheumatology, Taiyuan, Shanxi, China
| |
Collapse
|
3
|
Ming B, Li L, Cai S, Hu Z, Gao R, Umehara H, Zhong J, Zheng F, Dong L. How to focus on autoantigen-specific lymphocytes: a review on diagnosis and treatment of Sjogren's syndrome. J Leukoc Biol 2025; 117:qiae247. [PMID: 39953919 DOI: 10.1093/jleuko/qiae247] [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: 06/18/2024] [Revised: 08/30/2024] [Indexed: 02/17/2025] Open
Abstract
Sjogren's syndrome (SS) is an autoimmune epithelitis characterized by focal lymphocytic infiltration against self-antigens leading to progressive glandular dysfunction, which can develop to multisystem manifestation. The classification criteria for SS emphasizes glandular lymphocyte infiltrates and anti-SSA/SSB seropositivity, which is usually manifested in advanced patients. Therapeutically, apart from symptomatic treatment, treatment of SS is based on glucocorticoids and conventional synthetic disease-modifying antirheumatic drugs with global immunosuppression, but the efficacy of biologic or targeted synthetic therapies is still sparse. Currently, emerging studies focus on autoantigen-specific immunotherapies to treat autoimmune disorders by directly eliminating autoreactive cell subsets and inducing tolerance by increasing the autoreactive regulatory lymphocytes. Herein, we summarize the current state of research on the autoantigen-specific approaches for detecting autoreactive lymphocytes and outline the current autoantigen-specific immunotherapies in other autoimmune disorders and their attempts in treatment of SS. Last, we discuss the potential value of focusing on autoantigen-specific lymphocytes in the early diagnosis, monitoring, and targeted treatment of SS. Potential strategies for targeting autoreactive lymphocytes need to be confirmed in SS.
Collapse
Affiliation(s)
- Bingxia Ming
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling Li
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shaozhe Cai
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ziwei Hu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Rongfen Gao
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hisanori Umehara
- Department of Medicine, Nagahama City Hospital, Nagahama 526-0043, Japan
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei 430030, China
- NHC Key Laboratory of Organ Transplantation, Wuhan, Hubei 430030, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
4
|
Wu D, Yin M, Cao D, Zhang X, Zhu Y, Wei Y, Li Y, Wen C, Zhou J. Disruption of Gut Microbiota and Associated Fecal Metabolites in Collagen-Induced Arthritis Mice During the Early Stage. J Inflamm Res 2025; 18:1703-1717. [PMID: 39925933 PMCID: PMC11806705 DOI: 10.2147/jir.s502980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease and increasing evidence suggests that disturbances in the composition and function of gut microbiota are potentially implicated in the progression of RA. Further revealing the microbiota and related metabolic disorders in the preclinical stage of RA (pre-RA) is of great significance for exploration of disease mechanisms. Methods DBA/1 mice were injected with type II collagen on days 0 and 21 to establish collagen-induced arthritis (CIA) mouse model. Footpad thickness, serum autoantibodies, and joint histopathology were used to assess the progression of RA. A combination of 16S rRNA sequencing, untargeted metabolomics and targeted short-chain fatty acids (SCFAs) analysis were employed to comprehensively investigate the alterations of gut microbiota and fecal metabolites in CIA during the pre-RA stage. Results 20 days after the initial collagen immunization, CIA mice showed immune responses without joint symptoms, alongside gut microbiota disruption. Alterations were observed in 20 microbial taxa, including Oscillospira, Bifidobacterium, Ruminococcus, Allobaculum, Alistipes, Lactobacillus, and Candidatus_Arthromitus, etc. Untargeted and targeted metabolomics identified 33 altered fecal metabolites, mainly including sugars and their derivatives, amino acids, long-chain fatty acids and SCFAs, etc. Correlation analysis showed significant correlations between specific gut microbial abundances and fecal metabolite levels. Especially, SCFAs were strongly associated with Bifidobacterium, Alistipes, Ruminococcus, Anaerotruncus, and Allobaculum. Conclusion These findings suggest that collagen immunization leads to disruption of gut microbiome and induces changes of fecal metabolites in mice, which may play a key role in early development of RA in CIA mice.
Collapse
Affiliation(s)
- Dehong Wu
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, People’s Republic of China
| | - Mengdi Yin
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Dandan Cao
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Xiafeng Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yichun Zhu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Ying Wei
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yiling Li
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Chengping Wen
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Jia Zhou
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
5
|
Zhou J, Liu Y, Wu J. Association between immune cells, inflammatory cytokines, and sarcopenia: Insights from a Mendelian randomization analysis. Arch Gerontol Geriatr 2025; 128:105560. [PMID: 39213747 DOI: 10.1016/j.archger.2024.105560] [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: 04/06/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Recent studies have suggested a possible link between sarcopenia, immune dysregulation, and chronic inflammation, although the specific immune components implicated remain unclear. This investigation employs Mendelian Randomization (MR) to explore the reciprocal relationship between immune cells, inflammatory markers, and sarcopenia. METHOD We performed two-sample and multivariate MR analyses using publicly accessible genome-wide association studies (GWAS) summary statistics. Our analyses included 731 immune cells, 41 inflammatory cytokines, and sarcopenia related traits (appendicular lean mass [ALM], low hand-grip strength [LHS], and walking pace [WP]), with additional sensitivity analyses conducted to confirm the findings. RESULTS After false discovery rate (FDR) correction, significant associations were found between ten immune traits and ALM, with the CD127 marker in the Treg panel showing consistent positive correlation across four sites. In contrast, NKT%lymphocyte negatively correlated with WP (OR = 0.99, P = 0.023). In terms of inflammatory cytokines, macrophage colony-stimulating factor (MCSF) (OR = 1.03, P = 0.024) and hepatocyte growth factor (HGF) (OR = 1.03, P = 0.002) demonstrated positive associations with ALM, while interleukin-16 (IL-16) (OR = 0.99, P = 0.006) was inversely related. The reverse Mendelian randomization analysis found no direct causal links between sarcopenia traits and immune or inflammatory markers. Sensitivity analyses underscored the findings' resilience to pleiotropy, and adjusting for inter-trait dynamics weakened these relationships in the multivariable MR analysis. CONCLUSION Our study reveals causal associations between specific immune phenotypes, inflammatory cytokines, and sarcopenia, providing insight into the development of sarcopenia and potential treatment strategies.
Collapse
Affiliation(s)
- Jinqiu Zhou
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Liu
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinhui Wu
- Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Yuan L, Li Y, Liu D, Zhang H, Yang J, Shen H, Xia L, Yao L, Lu J. Interleukin-35 protein inhibits osteoclastogenesis and attenuates collagen-induced arthritis in mice. J Cell Physiol 2024; 239:e31231. [PMID: 38451477 DOI: 10.1002/jcp.31231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 03/08/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease. Its pathological features include synovial inflammation, bone erosion, and joint structural damage. Our previous studies have shown that interleukin (IL)-35 is involved in the pathogenesis of bone loss in RA patients. In this study, we are further evaluating the efficacy of IL-35 on collagen-induced arthritis (CIA) in the mouse model. Male DBA/1J mice (n = 10) were initially immunized, 2 μg/mouse IL-35 was injected intraperitoneally every week for 3 weeks after the establishment of the CIA model. Clinical arthritis, histopathological analysis, and three-dimensional micro‑computed tomography (3D micro‑CT) were determined after the mice were anesthetized on the 42th day. In vitro, RANKL/M-CSF induced mouse preosteoclasts (RAW264.7 cells line) was subjected to antiarthritis mechanism study in the presence of IL-35. The results of clinical arthritis, histopathological analysis, and 3D micro‑CT, the expression of RANK/RANKL/OPG axis, inflammatory cytokines, and osteoclastogenesis-related makers demonstrated decreasing severity of synovitis and bone destruction in the ankle joints after IL-35 treatment. Furthermore, IL-35 attenuated inflammatory cytokine production and the expression of osteoclastogenesis-related makers in a mouse preosteoclasts cell line RAW264.7. The osteoclastogenesis-related makers were significantly reduced in IL-35 treated RAW264.7 cells line after blockage with the JAK/STAT1 signaling pathway. These results demonstrated that IL-35 protein could inhibits osteoclastogenesis and attenuates CIA in mice. We concluded that IL-35 can exhibit anti-osteoclastogenesis effects by reducing the expression of inflammatory cytokines and osteoclastogenesis-related makers, thus alleviating bone destruction in the ankle joint and could be a potential therapeutic target for RA.
Collapse
Affiliation(s)
- Lin Yuan
- Department of Health Management, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yuxuan Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dan Liu
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Zhang
- Department of Rheumatology and Immunology, The Fifth People Hospital, Shenyang, China
| | - Jie Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hui Shen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Liping Xia
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lutian Yao
- Department of Orthopedic, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jing Lu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Patil SJ, Thorat VM, Koparde AA, Bhinge SD, Chavan DD, Bhosale RR. Unlocking the Future: New Biologic Therapies for Rheumatoid Arthritis. Cureus 2024; 16:e72486. [PMID: 39600762 PMCID: PMC11592031 DOI: 10.7759/cureus.72486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disorder that leads to joint destruction and functional disability. Traditional treatments, including disease-modifying antirheumatic drugs (DMARDs), often fail, leaving many patients without remission. The advent of biologic therapies that target specific immune system components (e.g., cytokines, T cells) has transformed RA treatment by offering new management options. These biologics (e.g., TNF inhibitors, interleukin blockers) are highly effective in controlling disease activity and preventing joint destruction. However, their use comes with safety concerns, particularly regarding immunosuppression and infection risks. Although still experimental, studies predict that future research will focus on enhancing the clinical response and safety of these agents through personalized approaches or novel mechanisms of action.
Collapse
Affiliation(s)
- Sarika J Patil
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Vandana M Thorat
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Akshada A Koparde
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Somnath D Bhinge
- Department of Pharmaceutical Chemistry, Rajarambapu College of Pharmacy, Kasegaon, IND
| | - Dhanashri D Chavan
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Rohit R Bhosale
- Department of Pharmaceutics, Krishna Foundation's Jaywant Institute of Pharmacy, Wathar, IND
| |
Collapse
|
8
|
Pružinská K, Chrastina M, Khademnematolahi S, Vyletelová V, Gajdošová L, Pastvová L, Dráfi F, Poništ S, Pašková Ľ, Kucharská J, Sumbalová Z, Muchová J, Martiniaková S, Bauerová K. Astaxanthin, Compared to Other Carotenoids, Increases the Efficacy of Methotrexate in Rat Adjuvant Arthritis. Int J Mol Sci 2024; 25:8710. [PMID: 39201397 PMCID: PMC11354740 DOI: 10.3390/ijms25168710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
This in vivo study performed in rat adjuvant arthritis aims to advance the understanding of astaxanthin's therapeutic properties for the possible treatment of rheumatoid arthritis (RA) in monotherapy and along with the standard RA treatment, methotrexate (MTX), in combination therapy. The main goal was to elucidate astaxanthin's full therapeutic potential, evaluate its dose dependency, and compare its effects in monotherapy with other carotenoids such as β-carotene and β-cryptoxanthin (KXAN). Moreover, potential differences in therapeutic activity caused by using different sources of astaxanthin, synthetic (ASYN) versus isolated from Blakeslea trispora (ASTAP), were evaluated using one-way ANOVA (Tukey-Kramer post hoc test). KXAN was the most effective in reducing plasma MMP-9 levels in monotherapy, significantly better than MTX, and in reducing hind paw swelling. The differences in the action of ASTAP and ASYN have been observed across various biometric, anti-inflammatory, and antioxidative parameters. In combined therapy with MTX, the ASYN + MTX combination proved to be better. These findings, especially the significant anti-arthritic effect of KXAN and ASYN + MTX, could be the basis for further preclinical studies.
Collapse
Affiliation(s)
- Katarína Pružinská
- Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Malá Hora 10701/4A, 036 01 Martin, Slovakia;
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS, 841 04 Bratislava, Slovakia; (M.C.); (S.K.); (F.D.); (S.P.)
| | - Martin Chrastina
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS, 841 04 Bratislava, Slovakia; (M.C.); (S.K.); (F.D.); (S.P.)
| | - Sasan Khademnematolahi
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS, 841 04 Bratislava, Slovakia; (M.C.); (S.K.); (F.D.); (S.P.)
- Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Veronika Vyletelová
- Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (V.V.); (Ľ.P.)
| | - Lívia Gajdošová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Sasinkova 2, 813 72 Bratislava, Slovakia; (L.G.); (L.P.); (Z.S.); (J.M.)
| | - Lucia Pastvová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Sasinkova 2, 813 72 Bratislava, Slovakia; (L.G.); (L.P.); (Z.S.); (J.M.)
| | - František Dráfi
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS, 841 04 Bratislava, Slovakia; (M.C.); (S.K.); (F.D.); (S.P.)
| | - Silvester Poništ
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS, 841 04 Bratislava, Slovakia; (M.C.); (S.K.); (F.D.); (S.P.)
| | - Ľudmila Pašková
- Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (V.V.); (Ľ.P.)
| | - Jarmila Kucharská
- Pharmacobiochemical Laboratory of Third Department of Internal Medicine, Faculty of Medicine, Comenius University in Bratislava, Špitálska 24, 813 72 Bratislava, Slovakia;
| | - Zuzana Sumbalová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Sasinkova 2, 813 72 Bratislava, Slovakia; (L.G.); (L.P.); (Z.S.); (J.M.)
| | - Jana Muchová
- Institute of Medical Chemistry, Biochemistry and Clinical Biochemistry, Faculty of Medicine, Comenius University, Sasinkova 2, 813 72 Bratislava, Slovakia; (L.G.); (L.P.); (Z.S.); (J.M.)
| | - Silvia Martiniaková
- Department of Food Technology, Institute of Food Science and Nutrition, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, 812 37 Bratislava, Slovakia;
| | - Katarína Bauerová
- Institute of Experimental Pharmacology and Toxicology, Centre of Experimental Medicine SAS, 841 04 Bratislava, Slovakia; (M.C.); (S.K.); (F.D.); (S.P.)
| |
Collapse
|
9
|
An X, Yang J, Cui X, Zhao J, Jiang C, Tang M, Dong Y, Lin L, Li H, Wang F. Advances in local drug delivery technologies for improved rheumatoid arthritis therapy. Adv Drug Deliv Rev 2024; 209:115325. [PMID: 38670229 DOI: 10.1016/j.addr.2024.115325] [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: 11/03/2023] [Revised: 03/25/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease characterized by an inflammatory microenvironment and cartilage erosion within the joint cavity. Currently, antirheumatic agents yield significant outcomes in RA treatment. However, their systemic administration is limited by inadequate drug retention in lesion areas and non-specific tissue distribution, reducing efficacy and increasing risks such as infection due to systemic immunosuppression. Development in local drug delivery technologies, such as nanostructure-based and scaffold-assisted delivery platforms, facilitate enhanced drug accumulation at the target site, controlled drug release, extended duration of the drug action, reduced both dosage and administration frequency, and ultimately improve therapeutic outcomes with minimized damage to healthy tissues. In this review, we introduced pathogenesis and clinically used therapeutic agents for RA, comprehensively summarized locally administered nanostructure-based and scaffold-assisted drug delivery systems, aiming at improving the therapeutic efficiency of RA by alleviating the inflammatory response, preventing bone erosion and promoting cartilage regeneration. In addition, the challenges and future prospects of local delivery for clinical translation in RA are discussed.
Collapse
Affiliation(s)
- Xiaoran An
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jiapei Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Xiaolin Cui
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Jiaxuan Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Chenwei Jiang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Minglu Tang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Yabing Dong
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Longfei Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Hui Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330000, PR China
| | - Feihu Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China.
| |
Collapse
|
10
|
Triaille C, Quartier P, De Somer L, Durez P, Lauwerys BR, Verschueren P, Taylor PC, Wouters C. Patterns and determinants of response to novel therapies in juvenile and adult-onset polyarthritis. Rheumatology (Oxford) 2024; 63:594-607. [PMID: 37725352 PMCID: PMC10907821 DOI: 10.1093/rheumatology/kead490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
Biologic and targeted synthetic DMARDs (b/tsDMARDs) have revolutionized the management of multiple rheumatic inflammatory conditions. Among these, polyarticular JIA (pJIA) and RA display similarities in terms of disease pathophysiology and response pattern to b/tsDMARDs. Indeed, the therapeutic efficacy of novel targeted drugs is variable among individual patients, in both RA and pJIA. The mechanisms and determinants of this heterogeneous response are diverse and complex, such that the development of true 'precision'-medicine strategies has proven highly challenging. In this review, we will discuss pathophysiological, patient-specific, drug-specific and environmental factors contributing to individual therapeutic response in pJIA in comparison with what is known in RA. Although some biomarkers have been identified that stratify with respect to the likelihood of either therapeutic response or non-response, few have proved useful in clinical practice so far, likely due to the complexity of treatment-response mechanisms. Consequently, we propose a pragmatic, patient-centred and clinically based approach, i.e. personalized instead of biomarker-based precision medicine in JIA.
Collapse
Affiliation(s)
- Clément Triaille
- Pôle de Pathologies Rhumatismales Systémiques et Inflammatoires, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Pediatric Hematology, Oncology, Immunology and Rheumatology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Division of Pediatric Rheumatology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Pierre Quartier
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
- Université Paris-Cité, Paris, France
- Member of the European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases – Project ID No. 739543
| | - Lien De Somer
- Division of Pediatric Rheumatology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Member of the European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases – Project ID No. 739543
- Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Patrick Durez
- Pôle de Pathologies Rhumatismales Systémiques et Inflammatoires, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Rheumatology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bernard R Lauwerys
- Pôle de Pathologies Rhumatismales Systémiques et Inflammatoires, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Patrick Verschueren
- Member of the European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases – Project ID No. 739543
- Department of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Carine Wouters
- Division of Pediatric Rheumatology, Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France
- Member of the European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases – Project ID No. 739543
- Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|
11
|
Martínez-Ramos S, García S. An update of murine models and their methodologies in immune-mediated joint damage and pain research. Int Immunopharmacol 2024; 128:111440. [PMID: 38176343 DOI: 10.1016/j.intimp.2023.111440] [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: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Murine models have played an indispensable role in the understanding of rheumatic and musculoskeletal disorders (RMD), elucidating the genetic, endocrine and biomechanical pathways involved in joint pathology and associated pain. To date, the available models in RMD can be classified as induced or spontaneous, both incorporating transgenic alternatives that improve specific insights. It is worth noting that the selection of the most appropriate model together with the evaluation of their specific characteristics and technical capabilities are crucial when designing the experiments. Furthermore, it is also imperative to consistently adhere to the ethical standards concerning animal experimentation. Recognizing the inherent limitation that any model can entirely encapsulates the complexity of the pathophysiology of these conditions, the aim of this review is to provide an updated overview on the methodology of current murine models in major arthropathies and their immune-mediated pathways, addressing to basic, translational and pharmacological research in joint damage and pain.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain.
| | - Samuel García
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| |
Collapse
|
12
|
Zeng L, Yu G, Yang K, He Q, Hao W, Xiang W, Long Z, Chen H, Tang X, Sun L. Exploring the mechanism of Celastrol in the treatment of rheumatoid arthritis based on systems pharmacology and multi-omics. Sci Rep 2024; 14:1604. [PMID: 38238321 PMCID: PMC10796403 DOI: 10.1038/s41598-023-48248-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 11/23/2023] [Indexed: 01/22/2024] Open
Abstract
To explore the molecular network mechanism of Celastrol in the treatment of rheumatoid arthritis (RA) based on a novel strategy (integrated systems pharmacology, proteomics, transcriptomics and single-cell transcriptomics). Firstly, the potential targets of Celastrol and RA genes were predicted through the database, and the Celastrol-RA targets were obtained by taking the intersection. Then, transcriptomic data and proteomic data of Celastrol treatment of RA were collected. Subsequently, Celastrol-RA targets, differentially expressed genes, and differentially expressed proteins were imported into Metascape for enrichment analysis, and related networks were constructed. Finally, the core targets of Celastrol-RA targets, differentially expressed genes, and differentially expressed proteins were mapped to synoviocytes of RA mice to find potential cell populations for Celastrol therapy. A total of 195 Celastrol-RA targets, 2068 differential genes, 294 differential proteins were obtained. The results of enrichment analysis showed that these targets, genes and proteins were mainly related to extracellular matrix organization, TGF-β signaling pathway, etc. The results of single cell sequencing showed that the main clusters of these targets, genes, and proteins could be mapped to RA synovial cells. For example, Mmp9 was mainly distributed in Hematopoietic cells, especially in Ptprn+fibroblast. The results of molecular docking also suggested that Celastrol could stably combine with molecules predicted by network pharmacology. In conclusion, this study used systems pharmacology, transcriptomics, proteomics, single-cell transcriptomics to reveal that Celastrol may regulate the PI3K/AKT signaling pathway by regulating key targets such as TNF and IL6, and then play an immune regulatory role.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qi He
- Department of Rehabilitation Medicine, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Wensa Hao
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wang Xiang
- Department of Rheumatology, The First People's Hospital Changde City, Changde, China
| | - Zhiyong Long
- Department of Rehabilitation Medicine, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Hua Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Anhui, China.
| |
Collapse
|
13
|
Kim A, Xie F, Abed OA, Moon JJ. Vaccines for immune tolerance against autoimmune disease. Adv Drug Deliv Rev 2023; 203:115140. [PMID: 37980949 PMCID: PMC10757742 DOI: 10.1016/j.addr.2023.115140] [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: 09/22/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The high prevalence and rising incidence of autoimmune diseases have become a prominent public health issue. Autoimmune disorders result from the immune system erroneously attacking the body's own healthy cells and tissues, causing persistent inflammation, tissue injury, and impaired organ function. Existing treatments primarily rely on broad immunosuppression, leaving patients vulnerable to infections and necessitating lifelong treatments. To address these unmet needs, an emerging frontier of vaccine development aims to restore immune equilibrium by inducing immune tolerance to autoantigens, offering a potential avenue for a cure rather than mere symptom management. We discuss this burgeoning field of vaccine development against inflammation and autoimmune diseases, with a focus on common autoimmune disorders, including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, inflammatory bowel disease, and systemic lupus erythematosus. Vaccine-based strategies provide a new pathway for the future of autoimmune disease therapeutics, heralding a new era in the battle against inflammation and autoimmunity.
Collapse
Affiliation(s)
- April Kim
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fang Xie
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Omar A Abed
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor 48109, USA.
| |
Collapse
|
14
|
Alshehri S, AlGhamdi SA, Alghamdi AM, Imam SS, Mahdi WA, Almaniea MA, Hajjar BM, Al-Abbasi FA, Sayyed N, Kazmi I. Protective effect of fustin against adjuvant-induced arthritis through the restoration of proinflammatory response and oxidative stress. PeerJ 2023; 11:e15532. [PMID: 37520245 PMCID: PMC10386820 DOI: 10.7717/peerj.15532] [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: 01/18/2023] [Accepted: 05/19/2023] [Indexed: 08/01/2023] Open
Abstract
Rheumatoid arthritis causes irreparable damage to joints. The present research sought to check fustin's anti-arthritic efficacy against the complete Freund's adjuvant-induced arthritis paradigm in animals by altering the inflammatory response. In the rats, complete Freund's adjuvant was used to trigger arthritis and they received fustin at 50 and 100 mg/kg for 21 days. At regular intervals, the hind paw volume and arthritic score were assessed. After the trial period, hematological, antioxidant, pro-inflammatory cytokines, and other biochemical parameters were estimated. Fustin-treated rats showed the down-regulation of hind paw volume, arthritic score, and altered hematological parameters (TLC, DLC (neutrophil, lymphocyte, monocyte, eosinophil, basophil)). Furthermore, fustin significantly mitigates proinflammatory cytokine (reduced interleukin, tumor necrosis factor-a (TNF-α), IL-6, IL-1β), oxidative stress (attenuated malondialdehyde (MDA), catalase (CAT), glutathione (GSH), superoxide dismutase (SOD)), attenuated production of prostaglandin E2 and myeloperoxidase (MPO) and improved nuclear factor erythroid 2-related factor (Nrf2) action. Fustin led to the benefit in arthritis-prone animals elicited by complete Freund's adjuvant via pro-inflammatory cytokine.
Collapse
Affiliation(s)
- Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shareefa A. AlGhamdi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amira M. Alghamdi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wael A. Mahdi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad A. Almaniea
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Baraa Mohammed Hajjar
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nadeem Sayyed
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
Fu W, Liu CJ. Unraveling the mechanisms behind joint damage. eLife 2023; 12:e89778. [PMID: 37366155 PMCID: PMC10299819 DOI: 10.7554/elife.89778] [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] [Indexed: 06/28/2023] Open
Abstract
A subtype of myeloid monocyte mediates the transition from autoimmunity to joint destruction in rheumatoid arthritis.
Collapse
Affiliation(s)
- Wenyu Fu
- Department of Orthopaedic Surgery, New York University Grossman SchoolNew YorkUnited States
- Department of Orthopaedics and Rehabilitation, Yale University School of MedicineNew HavenUnited States
| | - Chuan-ju Liu
- Department of Orthopaedic Surgery, New York University Grossman SchoolNew YorkUnited States
- Department of Orthopaedics and Rehabilitation, Yale University School of MedicineNew HavenUnited States
| |
Collapse
|
16
|
Su R, Li B, Wu R, Xie Y, Gao A, Gao C, Li X, Wang C. Stratified distribution of Th17 and Treg cells in patients with multi-stage rheumatoid arthritis. Arthritis Res Ther 2023; 25:55. [PMID: 37016395 PMCID: PMC10071616 DOI: 10.1186/s13075-023-03041-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/29/2023] [Indexed: 04/06/2023] Open
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a typical, progressive autoimmune disease. Its occurrence and development are associated with dysregulation of T and B cell numbers. However, the specific immune characteristics of different RA courses remain incompletely defined. Here, we describe the peripheral blood lymphocyte subsets, particularly CD4 + T subsets, of different RA courses with a focus on early RA (Ea-RA). METHODS In all, 131 patients with Ea-RA, 117 with advanced RA (Ad-RA), and 109 with treated RA (Tr-RA) were enrolled. We collected general clinical data. Whole blood samples obtained from the patients and 97 healthy controls (HCs) were analysed via flow cytometry. RESULTS Decreased absolute NK cell numbers and increased CD4/CD8 T cell ratios were observed in different RA groups, including Ea-RA, compared to healthy controls. In Ea-RA patients, the Th17 and Treg cell numbers were similar to those in HCs. We performed k-means clustering based on the profiles of Th17 and Treg cells for patients with multi-stage of RA. We identified three patient types: type A characterised by relatively low Treg and Th17 cell numbers, type B with moderate levels of Treg cells and levels of Th17 cells similar to that of type C patients, and type C with high levels of Treg cells and levels of Th17 cells similar to that of type B patients. CONCLUSION The immune characteristics of Ea-RA patients differ from those of HCs; an immune system disorder is apparent although no differences in Th17 and Treg levels were evident between Ea-RA patients and HCs. We found distributional heterogeneities of Th17 and Treg cells in patients with multi-stage of RA. Stratified management based on such heterogeneity may serve as a useful novel immunotherapy allowing of early intervention.
Collapse
Affiliation(s)
- Rui Su
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Ruihe Wu
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Yuhuan Xie
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Anqi Gao
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Brigham and Women's Hospital/Children's Hospital Boston, Joint Program in Transfusion Medicine, Harvard Medical School, PathologyBoston, USA
| | - Xiaofeng Li
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, the Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China.
| |
Collapse
|
17
|
Remodeling articular immune homeostasis with an efferocytosis-informed nanoimitator mitigates rheumatoid arthritis in mice. Nat Commun 2023; 14:817. [PMID: 36781864 PMCID: PMC9925448 DOI: 10.1038/s41467-023-36468-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/02/2023] [Indexed: 02/15/2023] Open
Abstract
Massive intra-articular infiltration of proinflammatory macrophages is a prominent feature of rheumatoid arthritis (RA) lesions, which are thought to underlie articular immune dysfunction, severe synovitis and ultimately joint erosion. Here we report an efferocytosis-informed nanoimitator (EINI) for in situ targeted reprogramming of synovial inflammatory macrophages (SIMs) that thwarts their autoimmune attack and reestablishes articular immune homeostasis, which mitigates RA. The EINI consists of a drug-based core with an oxidative stress-responsive phosphatidylserine (PtdSer) corona and a shell composed of a P-selectin-blocking motif, low molecular weight heparin (LMWH). When systemically administered, the LMWH on the EINI first binds to P-selectin overexpressed on the endothelium in subsynovial capillaries, which functions as an antagonist, disrupting neutrophil synovial trafficking. Due to the strong dysregulation of the synovial microvasculature, the EINI is subsequently enriched in the joint synovium where the shell is disassembled upon the reactive oxygen species stimulation, and PtdSer corona is then exposed. In an efferocytosis-like manner, the PtdSer-coroneted core is in turn phagocytosed by SIMs, which synergistically terminate SIM-initiated pathological cascades and serially reestablish intra-articular immune homeostasis, conferring a chondroprotective effect. These findings demonstrate that SIMs can be precisely remodeled via the efferocytosis-mimetic strategy, which holds potential for RA treatment.
Collapse
|
18
|
Corneth OBJ, Neys SFH, Hendriks RW. Aberrant B Cell Signaling in Autoimmune Diseases. Cells 2022; 11:cells11213391. [PMID: 36359789 PMCID: PMC9654300 DOI: 10.3390/cells11213391] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
Abstract
Aberrant B cell signaling plays a critical in role in various systemic and organ-specific autoimmune diseases. This is supported by genetic evidence by many functional studies in B cells from patients or specific animal models and by the observed efficacy of small-molecule inhibitors. In this review, we first discuss key signal transduction pathways downstream of the B cell receptor (BCR) that ensure that autoreactive B cells are removed from the repertoire or functionally silenced. We provide an overview of aberrant BCR signaling that is associated with inappropriate B cell repertoire selection and activation or survival of peripheral B cell populations and plasma cells, finally leading to autoantibody formation. Next to BCR signaling, abnormalities in other signal transduction pathways have been implicated in autoimmune disease. These include reduced activity of several phosphates that are downstream of co-inhibitory receptors on B cells and increased levels of BAFF and APRIL, which support survival of B cells and plasma cells. Importantly, pathogenic synergy of the BCR and Toll-like receptors (TLR), which can be activated by endogenous ligands, such as self-nucleic acids, has been shown to enhance autoimmunity. Finally, we will briefly discuss therapeutic strategies for autoimmune disease based on interfering with signal transduction in B cells.
Collapse
|
19
|
Bu Y, Wu H, Deng R, Wang Y. Geniposide restricts angiogenesis in experimentary arthritis via inhibiting Dnmt1-mediated PTEN hypermethylation. Int Immunopharmacol 2022; 111:109087. [PMID: 35908504 DOI: 10.1016/j.intimp.2022.109087] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/01/2022]
Abstract
Neovascularization in rheumatoid arthritis (RA) is a key bridge between malignant proliferative synovial tissue and pannus. In view of previous studies on the efficacy of Geniposide (GE) in experimentary arthritis, the purpose of this study was to investigate the possible mechanism of GE inhibiting angiogenesis by regulating the gene of phosphate and tension homology deleted on chromosome ten (PTEN). In this study, human umbilical vein endothelial cells (HUVEC) and adjuvant arthritis (AA) rat models were performed to research in vitro and in vivo. The results showed that GE treatment significantly reduced synovitis and angiogenesis in AA rats, which may be associated with the increased expression of PTEN with GE treatment. Meanwhile, the hypermethylation of PTEN accompanied by the over-expression of DNA methyltransferases (Dnmts) was demonstrated in TNF-α-induced HUVEC and AA rats. Knockdown of Dnmt1 by Dnmt1- siRNA significantly inhibited the tube formation of HUVEC in vitro. GE significantly restricted the angiogenesis of HUVEC by inhibiting DNA methylation, which was attributed to the down-regulation of Dnmt1 rather than Dnmt3a and Dnmt3b. The anti-angiogenesis effect of GE was further verified in AA model by the inhibition of Dnmt1. These results indicate that GE exhibited anti-angiogenesis effects in experimentary arthritis by inhibiting Dnmt1-mediated PTEN gene hypermethylation, which may brings new insights for the prevention and research of RA.
Collapse
Affiliation(s)
- Yanhong Bu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China.
| | - Ran Deng
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Yan Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
20
|
Immune system and sarcopenia: Presented relationship and future perspective. Exp Gerontol 2022; 164:111823. [DOI: 10.1016/j.exger.2022.111823] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/29/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022]
|
21
|
Milchram L, Fischer A, Huber J, Soldo R, Sieghart D, Vierlinger K, Blüml S, Steiner G, Weinhäusel A. Functional Analysis of Autoantibody Signatures in Rheumatoid Arthritis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041452. [PMID: 35209238 PMCID: PMC8876797 DOI: 10.3390/molecules27041452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 11/21/2022]
Abstract
For the identification of antigenic protein biomarkers for rheumatoid arthritis (RA), we conducted IgG profiling on high density protein microarrays. Plasma IgG of 96 human samples (healthy controls, osteoarthritis, seropositive and seronegative RA, n = 24 each) and time-series plasma of a pristane-induced arthritis (PIA) rat model (n = 24 total) were probed on AIT’s 16k protein microarray. To investigate the analogy of underlying disease pathways, differential reactivity analysis was conducted. A total of n = 602 differentially reactive antigens (DIRAGs) at a significance cutoff of p < 0.05 were identified between seropositive and seronegative RA for the human samples. Correlation with the clinical disease activity index revealed an inverse correlation of antibodies against self-proteins found in pathways relevant for antigen presentation and immune regulation. The PIA model showed n = 1291 significant DIRAGs within acute disease. Significant DIRAGs for (I) seropositive, (II) seronegative and (III) PIA were subjected to the Reactome pathway browser which also revealed pathways relevant for antigen presentation and immune regulation; of these, seven overlapping pathways had high significance. We therefore conclude that the PIA model reflects the biological similarities of the disease pathogenesis. Our data show that protein array analysis can elucidate biological differences and pathways relevant in disease as well be a useful additional layer of omics information.
Collapse
Affiliation(s)
- Lisa Milchram
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria; (L.M.); (J.H.); (R.S.); (K.V.)
| | - Anita Fischer
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria; (A.F.); (D.S.); (S.B.); (G.S.)
| | - Jasmin Huber
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria; (L.M.); (J.H.); (R.S.); (K.V.)
| | - Regina Soldo
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria; (L.M.); (J.H.); (R.S.); (K.V.)
| | - Daniela Sieghart
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria; (A.F.); (D.S.); (S.B.); (G.S.)
| | - Klemens Vierlinger
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria; (L.M.); (J.H.); (R.S.); (K.V.)
| | - Stephan Blüml
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria; (A.F.); (D.S.); (S.B.); (G.S.)
| | - Günter Steiner
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria; (A.F.); (D.S.); (S.B.); (G.S.)
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Medical University of Vienna, Währinger Gürtel 18–20, 1090 Vienna, Austria
| | - Andreas Weinhäusel
- Center for Health and Bioresources, Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, Giefinggasse 4, 1210 Vienna, Austria; (L.M.); (J.H.); (R.S.); (K.V.)
- Correspondence:
| |
Collapse
|
22
|
Braun J, Westhoff T, Baraliakos X, Babel N. [Are SARS-CoV-2 reactive T cells detectable and potentially protective in patients under anti-CD20 treatment with an impaired humoral response?]. Z Rheumatol 2021; 80:879-883. [PMID: 34529110 PMCID: PMC8445008 DOI: 10.1007/s00393-021-01083-3] [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] [Accepted: 08/14/2021] [Indexed: 11/16/2022]
Abstract
The pandemic attributable to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has greatly changed life in most countries of the world for more than 1.5 years now. The spread could be more or less well-controlled and fatalities could partly be avoided by obligatory wearing of masks, contact restrictions and since the beginning of the year by vaccinations. Patients with chronic inflammatory diseases and organ transplant patients under immunosuppression, are somewhat more at risk to become ill with coronavirus disease 2019 (COVID-19). The probability and severity of an infection depends on the ability of the humoral and cellular immune systems to effectively combat the virus. This can be substantially improved by vaccination. The B cell depleting monoclonal antibody rituximab (RTX) is frequently employed in rheumatic diseases, whereby antibody formation against the new pathogen within the framework of vaccination is restricted. Recent study results in patients treated with RTX indicate that an effective cellular immune response can be developed despite the impaired humoral response.
Collapse
Affiliation(s)
- J Braun
- Rheumazentrum Ruhrgebiet Herne, Ruhr-Universität Bochum, Claudiusstr. 45, 44649, Herne, Deutschland.
| | - T Westhoff
- Medizinische Klinik I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Deutschland
| | - X Baraliakos
- Rheumazentrum Ruhrgebiet Herne, Ruhr-Universität Bochum, Claudiusstr. 45, 44649, Herne, Deutschland
| | - N Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Deutschland
- Berlin Center for Advanced Therapies (BeCAT), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Deutschland
| |
Collapse
|