1
|
Chen J, Ho CY, Tu YK, Lin YC, Hsia Y, Lin YC, Shantakumar S. A systematic review and meta-analysis of herpes zoster risk in adults with immunocompromised conditions and autoimmune diseases in Asia-Pacific. Hum Vaccin Immunother 2025; 21:2496048. [PMID: 40299930 PMCID: PMC12045574 DOI: 10.1080/21645515.2025.2496048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Asia-Pacific (APAC) faces an increasing burden of herpes zoster (HZ) over time. The risk of HZ and its complications are increased in immunocompromised (IC) patients and those with autoimmune diseases (AID). Our study aimed to synthesize evidence on the epidemiological burden of HZ and its complications among the general adult population and patients with IC/AID conditions in APAC. Following a systematic literature review, we performed meta-analyses for outcomes where ≥3 studies met the inclusion criteria. Of the 271 articles identified, 75 were included for meta-analysis. We found a high burden of HZ and its complications (i.e., postherpetic neuralgia, HZ ophthalmicus), particularly among individuals with IC/AID conditions in APAC. Patients with IC/AID conditions had significantly increased HZ risk and a higher proportion of HZ recurrence than the general adult population. These findings may inform clinical practice and public health decisions regarding HZ prevention, including HZ vaccination strategies, among the IC/AID population in APAC.
Collapse
Affiliation(s)
| | - Chin-Yen Ho
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Kang Tu
- Health Data Research Center, National Taiwan University, Taipei, Taiwan
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yi-Chih Lin
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Nephrology, National Taiwan University Hospital Jinshan Branch, New Taipei City, Taiwan
| | - Yun Hsia
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Chun Lin
- Institute of Epidemiology & Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Anesthesiology, MacKay Memorial Hospital, Taipei, Taiwan
| | | |
Collapse
|
2
|
Zeng X, Gao Y, Bahabayi A, Alimu X, Liu T, Zheng M, Zhang Z, Li Q, Liu C. Upregulated TCF1 + Treg Cells With Stronger Function in Systemic Lupus Erythematosus Through Activation of the Wnt-β-Catenin. Immunology 2025; 175:251-262. [PMID: 40129177 DOI: 10.1111/imm.13914] [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: 08/16/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 03/26/2025] Open
Abstract
The role of T-cell factor 1 (TCF1) in human regulatory T cells (Treg) and its clinical significance in systemic lupus erythematosus (SLE) remain unclear. Through bioinformatics analysis and flow cytometry, the Tcf7 gene and TCF1 protein were found to be highly expressed in Treg cells. TCF1+ Treg cells exhibited increased expression of CTLA4 and LAG3 and higher IL-10 secretion than TCF1- Treg cells. Circulating TCF1+ Treg cells were elevated and displayed increased inhibitory markers in SLE patients. The Wnt-β-catenin pathway was activated in TCF1+ Treg cells in SLE patients. The addition of XAV939 impaired the function of TCF1+ Treg cells. Clinically, TCF1+ Treg cells were not only related to CRP, ESR and IL-2, but also could differentiate SLE patients from healthy controls, primary Sjögren's syndrome patients and rheumatoid arthritis patients. In conclusion, the increased TCF1+ Treg cells in SLE patients indicate a stronger suppressive function for the activated Wnt-β-catenin pathway and help screening and assisting in the diagnosis of SLE patients.
Collapse
Affiliation(s)
- Xingyue Zeng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yiming Gao
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Xiayidan Alimu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Tianci Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Mohan Zheng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhonghui Zhang
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Qi Li
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| |
Collapse
|
3
|
Franco-Fuquen P, Figueroa-Aguirre J, Martínez DA, Moreno-Cortes EF, Garcia-Robledo JE, Vargas-Cely F, Castro-Martínez DA, Almaini M, Castro JE. Cellular therapies in rheumatic and musculoskeletal diseases. J Transl Autoimmun 2025; 10:100264. [PMID: 39931050 PMCID: PMC11808717 DOI: 10.1016/j.jtauto.2024.100264] [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: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 02/13/2025] Open
Abstract
A substantial proportion of patients diagnosed with rheumatologic and musculoskeletal diseases (RMDs) exhibit resistance to conventional therapies or experience recurrent symptoms. These diseases, which include autoimmune disorders such as multiple sclerosis, rheumatoid arthritis, and systemic lupus erythematosus, are marked by the presence of autoreactive B cells that play a critical role in their pathogenesis. The persistence of these autoreactive B cells within lymphatic organs and inflamed tissues impairs the effectiveness of B-cell-depleting monoclonal antibodies like rituximab. A promising therapeutic approach involves using T cells genetically engineered to express chimeric antigen receptors (CARs) that target specific antigens. This strategy has demonstrated efficacy in treating B-cell malignancies by achieving long-term depletion of malignant and normal B cells. Preliminary data from patients with RMDs, particularly those with lupus erythematosus and dermatomyositis, suggest that CAR T-cells targeting CD19 can induce rapid and sustained depletion of circulating B cells, leading to complete clinical and serological responses in cases that were previously unresponsive to conventional therapies. This review will provide an overview of the current state of preclinical and clinical studies on the use of CAR T-cells and other cellular therapies for RMDs. Additionally, it will explore potential future applications of these innovative treatment modalities for managing patients with refractory and recurrent manifestations of these diseases.
Collapse
Affiliation(s)
- Pedro Franco-Fuquen
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - David A. Martínez
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Eider F. Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan E. Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | | | - Mustafa Almaini
- Rheumatology, Allergy & Clinical Immunology Division, Mafraq Hospital, United Arab Emirates
| | - Januario E. Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA
- Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
4
|
Chen X, Shen B, Lin W, Xiong Z, Yang B, Luo H, Zong Z, Chen J, Bahabayi A, Liu C. Altered CD27-related T cell subsets reflect immune imbalance in systemic lupus erythematosus. Immunol Res 2025; 73:83. [PMID: 40381105 DOI: 10.1007/s12026-025-09637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
OBJECTIVE This study aims to analyze CD27 expression in various subsets of CD4+ T cells in peripheral blood, explore the functional characteristics of the CD27+ subsets in regulatory T cells (Tregs) and CD4+ T cells, and assess their immunological alterations in newly diagnosed systemic lupus erythematosus (SLE) patients. METHODS Peripheral blood was collected from 34 newly diagnosed, untreated SLE patients and 22 healthy controls. Flow cytometry was used to analyze CD27 expression on T cell subsets, comparing the functional markers between CD27+ and CD27- subsets. CD27 expression on Tregs and total CD4+ T cells in SLE patients and healthy controls were compared. ROC curves were constructed, and areas under the curve (AUCs) was calculated to evaluate the diagnostic value of CD27-related T cell subsets for SLE. RESULTS The proportion of Tregs in the peripheral blood of SLE patients was increased, and CD27 expression was higher in Tregs than in CD4+ T cells in healthy individuals. CD27+ CD4+ T cells exhibited higher CD45RA and lower CD226 expression. CD27+ Tregs showed higher Helios and TIGIT expression and lower CD226 expression. CD27+ cell proportions in both CD4+ T cells and Tregs were significantly reduced in SLE patients. The AUC for CD27-related T cell subsets in diagnosing newly diagnosed SLE ranged from 0.6238 to 0.86. CONCLUSION CD27+ CD4+ T cells show reduced activation features, while CD27+ Tregs exhibit enhanced regulatory potential. Their decreased proportions in SLE patients suggest early immune dysregulation.
Collapse
Affiliation(s)
- Xiaoning Chen
- Department of Clinical Laboratory, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Bing Shen
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Weijie Lin
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ziqi Xiong
- Department of Clinical Laboratory, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Bohao Yang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hanxi Luo
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhiwei Zong
- Institute of Medical Technology, Peking University Health Science Center, Beijing, China
| | - Jie Chen
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China
| | - Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, 11 Xizhimen South Street, Beijing, 100044, China.
| |
Collapse
|
5
|
Ji J, Yang S, Xu Y, He Q, Liang Q, Feng G, Xia Y, Yang M, Huang Y, Yang J, Dong C, Zhao R, Wang Y, Guo G, Sha X, Li J, Guo Y, Gu Z. M2-ApoBDs as a therapeutic strategy for systemic lupus erythematosus: targeted macrophage reprogramming and treg differentiation. J Nanobiotechnology 2025; 23:354. [PMID: 40380199 PMCID: PMC12083009 DOI: 10.1186/s12951-025-03437-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder that affects various organs and systems, significantly impacting patients' health and quality of life. Conventional drugs, including glucocorticoids and standard immunosuppressive drugs, may not be enough to achieve a satisfactory therapeutic outcome in some refractory SLE patients. The abnormal phenotype and function of macrophages participate in the development of SLE. The targeted delivery to reprogram macrophage in SLE has been a long-standing challenge. Apoptotic bodies (ApoBDs) are essential for intercellular communications. This study aims to explore an effective and targeted treatment to SLE via macrophage reprogramming and Treg differentiation. In this work, we found that M2 macrophages-derived ApoBDs (M2-ApoBDs) could selectively target and localize to the spleen, where they were engulfed by splenic macrophages (phagocytic rate 73.4%). Single-cell RNA sequencing revealed that the efferocytosis of M2-ApoBDs triggered transcriptional changes in M2 (anti-inflammatory) macrophages within the spleen, subsequently promoting the differentiation of Treg cells in vivo. Immunological experiments revealed that M2-ApoBDs prompted the reprogramming of M2 macrophages in vitro, which subsequently influenced Treg cell differentiation via ligand-receptor interactions. In SLE mice, M2-ApoBDs alleviated the disease progression, including 24-hours urinary protein, plasma creatinine, plasma C3 levels, and glomerular sclerosis and interstitial fibrosis. These findings show that M2-ApoBDs can targeted-modulate macrophage polarization and Treg immune regulation, offering a novel therapeutic strategy for the effective treatment of SLE.
Collapse
Affiliation(s)
- Juan Ji
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Shaoying Yang
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Yongxin Xu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Qian He
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Qian Liang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Guijuan Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Yunfei Xia
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Mei Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yuting Huang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Junling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Chen Dong
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Rui Zhao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yunan Wang
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Genkai Guo
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Xiaoqi Sha
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Jing Li
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| | - Yuehua Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China.
| |
Collapse
|
6
|
Martínez-Urbistondo M, Higuera-Gómez A, de Cuevillas B, Cuevas-Sierra A, Mellor-Pita S, Moreno-Torres V, Vargas JA, Castejón R, Martínez JA. Visceral fat, cardiovascular risk factors and quality of life in lupus activity categorised via complement C3. Lupus Sci Med 2025; 12:e001423. [PMID: 40374236 PMCID: PMC12083274 DOI: 10.1136/lupus-2024-001423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Patients with lupus face increased cardiovascular risk linked to their autoimmune status. This study assesses the relationships between cardiovascular risk factors, lifestyle and health-related quality of life (HRQoL) concerning SLE activity categorised by complement C3. METHODS 74 patients with SLE were recruited and stratified as active (C3 <90 mg/dL) or inactive (C3 >90 mg/dL), alongside 74 controls with obesity-related low-grade inflammation, at Hospital Universitario Puerta de Hierro Majadahonda. Anthropometric measurements, clinical and demographic data were recorded, and participants completed validated questionnaires on physical activity, dietary intake and HRQoL. Fasting blood samples were collected for metabolic determinations. Comparative analyses between SLE groups and controls, along with regression models adjusted for variables associated with disease activity, were performed. RESULTS The inactive SLE group exhibited a less healthy adiposity profile compared with the active group (36.7% vs 33.2% total fat mass; 8.5 AU vs 6.5 AU visceral fat mass) and showed a higher prevalence of cardiovascular risk factors, including markers of obesity, hypertension, dyslipidaemia and increased waist circumference, along with worse HRQoL outcomes. Notably, age, body mass index and insulin resistance were associated with SLE inactivity, while fibrinogen correlated with disease activity as assessed by complement C3 levels. Interestingly, household composition as a sociodemographic variable (alone, couple/children/elderly or other) also showed an independent association with SLE activity. CONCLUSIONS Inactive patients with SLE exhibited more adverse cardiovascular risk markers compared with active patients categorised by complement C3, even when glucocorticoid administration was accounted for. Additionally, this research highlights the potential influence of fibrinogen as well as metabolic and sociodemographic factors on disease activity. These findings emphasise the need for personalised precision management strategies such as measurement of fibrinogen levels and insulin resistance and sociodemographic considerations that address both cardiovascular risk and overall lifestyle plus exposome in patients with SLE and may partly explain SLE activity evolution.
Collapse
Affiliation(s)
| | - Andrea Higuera-Gómez
- Precision Nutrition and Cardiometabolic Health, IMDEA Food, Madrid, Spain
- Department of Pharmacy and Nutrition, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid - Campus de Villaviciosa de Odón, Madrid, Spain
| | | | | | - Susana Mellor-Pita
- Internal Medicine Service, Puerta de Hierro University Hospital of Majadahonda, Majadahonda, Spain
| | - Victor Moreno-Torres
- Puerta de Hierro University Hospital of Majadahonda, Majadahonda, Spain
- Health Sciences School and Medical Center, UNIR, Logrono, Spain
| | | | - Raquel Castejón
- Puerta de Hierro University Hospital of Majadahonda, Majadahonda, Spain
| | - J Alfredo Martínez
- Precision Nutrition and Cardiometabolic Health, IMDEA Food, Madrid, Spain
- Biomedical Research Centre for Obesity Physiopathology and Nutrition Network (CIBEROBN), Instituto de Salud Carlos III (ISCIII), CIBERON, Madrid, Spain
| |
Collapse
|
7
|
Lu RL, Chen YR, Yang XF, Huang XY, Liu DZ, Hong XP. Genetic liability to gut microbiota and inflammatory cytokines in relation to systemic lupus erythematosus risk: a multi-omics study. Clin Rheumatol 2025:10.1007/s10067-025-07435-7. [PMID: 40332703 DOI: 10.1007/s10067-025-07435-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES Systemic lupus erythematosus (SLE) has been associated with gut microbiota in some studies. There is no clear evidence that cytokines act as mediators. METHODS We first assessed the differences in gut microbiota between SLE patients and healthy controls using 16S rDNA sequencing. Subsequently, we used the summary statistics of gut microbiota, cytokines, and SLE from large genome-wide association studies. To explore the causal relationships between gut microbiota and SLE and identify potential mediating cytokines, we performed bidirectional Mendelian randomization analyses. Finally, the levels of potentially mediating cytokines were determined by ELISA. RESULTS Fecal 16S rDNA sequencing showed that there was gut microbiota disorder in SLE patients. Based on two-sample analysis, seven gut microbiota taxa were causally associated with SLE. SLE influenced the relative abundance of two gut microbiota taxa in our large-scale MR study. Mediation analyses revealed that the causal relationship between genus Lachnospiraceae UCG001 and SLE was exclusively mediated by fibroblast growth factor 19 (FGF19) levels and the causal relationship between order Lactobacillales and SLE was exclusively mediated by tumor necrosis factor receptor superfamily member 9 (TNFRSF9) levels. Elevated levels of FGF19 affected the association between the reduced relative abundance of the genus Coprobacter and SLE, mediating by a proportion of 10.64% (P = 0.030). Furthermore, ELISA showed that circulating TNFRSF9 and FGF19 levels were higher in SLE patients than healthy controls. CONCLUSION Our study demonstrated that there is a causal link between some gut microbiota taxa and SLE. In addition, we revealed possible mediating effects in this relationship. Key Points • We first demonstrate a causal association between gut microbiota, cytokines, and SLE comprehensively. • Our experiments also confirmed that TNFRSF9 and FGF19 may play a role in SLE. These results provide new ideas for microbiome-based investigation of new mechanisms and therapies for SLE.
Collapse
Affiliation(s)
- Rui-Ling Lu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Yan-Ran Chen
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Xu-Fa Yang
- Department of Rheumatology and Immunology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Xin-Yi Huang
- Department of Rheumatology and Immunology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China
| | - Dong-Zhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
- Department of Rheumatology and Immunology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| | - Xiao-Ping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
- Department of Rheumatology and Immunology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Shu J, Xie W, Mei C, Ren A, Ke S, Ma M, Zhou Z, Hu Y, Mei H. Safety and clinical efficacy of Relmacabtagene autoleucel (relma-cel) for systemic lupus erythematosus: a phase 1 open-label clinical trial. EClinicalMedicine 2025; 83:103229. [PMID: 40386685 PMCID: PMC12083988 DOI: 10.1016/j.eclinm.2025.103229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025] Open
Abstract
Background Systemic lupus erythematosus (SLE) is a classic systemic autoimmune disease mediated by autoantibodies. Chimeric antigen receptor T (CAR-T) cell therapy, known for its success in cancer, has shown promise in achieving durable B cell depletion and long-term remission in SLE. Relmacabtagene autoleucel (relma-cel) is the second anti-CD19 CAR-T product approved for marketing by the National Medical Products Administration (NMPA) in China and demonstrates its long-term efficacy in relapsed/refractory (r/r) large B cell lymphoma (LBCL). We report the results from a phase I open-label clinical trial of relma-cel in treating patients with moderately to severely active SLE. Methods Eligible patients were aged 18-70 years, a ≥6-month history of SLE, and the disease had to remain active after at least 2 months of stable SLE standard treatment prior to screening. We evaluated four dose levels (DL) of relma-cel in a dose-escalation scheme: total dose of 25 × 106, 50 × 106, 75 × 106, and 100 × 106 anti-CD19 CAR-T cells. All patients received lymphodepletion chemotherapy with fludarabine and cyclophosphamide. The primary endpoints were the incidence of dose-limiting toxicities (DLTs) and adverse events (AEs). Secondary endpoints included the evaluation of standard cellular pharmacokinetic parameters, the SLE Responder Index (SRI) response rate, and changes from baseline in the Safety of Estrogens in Lupus Erythematosus National Assessment-Systemic Lupus Erythematosus Disease Activity Index (SELENA-SLEDAI), British Isles Lupus Assessment Group 2004 (BILAG-2004) and Physician's Global Assessment (PGA) scores post-treatment. This trial is registered with ClinicalTrials.gov, NCT05765006. Findings Between March 28, 2023 and April 8, 2024, a total of 12 patients were screened for study inclusion, of whom 8 patients were enrolled and assigned to different dose levels: 25 × 106 cells (n = 3), 50 × 106 cells (n = 2), 75 × 106 cells (n = 2), and 100 × 106 cells (n = 1). No DLT was observed. The most common AEs included cytopenia (n = 8, 100%), cytokine release syndrome (CRS) (n = 7, 88%) and hypogammaglobulinemia (n = 5, 63%). No Grade 3 or higher immune effector cell-associated hematotoxicity (ICAHT) occurred. No cases of immune effector cell-associated neurotoxicity syndrome (ICANS) were reported. CRS was predominantly grade 1, characterized mainly by mild fever and muscle soreness. A rare severe adverse event, immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), was observed in one patient. The median time to reach maximum CAR-T cell expansion (Cmax) was 9.5 days (range: 8-22 days). The median Cmax was 18.74 CD3+CAR+ cells/μL (range: 7.94-228.36) by flow cytometry and 81766.5 copies/μg DNA (range: 50,979-1,140,893) by quantitative real-time PCR (qPCR). In all patients treated with relma-cel, CD19+ B cells in peripheral blood were almost completely depleted within 11-15 days and gradually recovered within 2-6 months. All patients achieved SRI response. Four patients achieved Definition of Remission in SLE (DORIS) remission criteria and seven patients reached the Lupus Low Disease Activity State (LLDAS) criteria within 1-4 months following relma-cel infusion. Interpretation This study preliminarily demonstrated that relma-cel is an effective and safe CAR-T product for the treatment of patients with moderately to severely active SLE, providing valuable clinical insights into the management of rare complications. Further studies with larger sample sizes are warranted. Funding National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Jinhui Shu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Wei Xie
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Chunli Mei
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anqi Ren
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Sha Ke
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Meilin Ma
- JW Therapeutics (Shanghai) Co. Ltd, Shanghai, China
| | - Zisong Zhou
- JW Therapeutics (Shanghai) Co. Ltd, Shanghai, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan, China
| |
Collapse
|
9
|
Qi B, Zhao J, Meng C, Bu P, Pu LQ, Zhao W, Li C, Xu Y. Exploring the association between systemic lupus erythematosus and osteonecrosis by Mendelian randomization analysis. Sci Rep 2025; 15:15040. [PMID: 40301378 PMCID: PMC12041352 DOI: 10.1038/s41598-025-95467-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/21/2025] [Indexed: 05/01/2025] Open
Abstract
Patients with systemic lupus erythematosus (SLE) have been shown to have a high risk of osteonecrosis, but the potential causal relationship between genetic susceptibility and risk of osteonecrosis is unclear. In this study, we used Mendelian randomization to investigate the effects of SLE, gout and rheumatoid arthritis on osteonecrosis, and performed post-GWAS localization and functional analyses of GWAS studies related to osteonecrosis, with the aim of obtaining a more in-depth understanding of the mechanisms of osteonecrosis. In this study, a total of 45 single nucleotide polymorphisms (SNPS) data associated with SLE from publicly available genome-wide association study (GIS) datasets were selected for magnetic resonance estimation using inverse-variance weighting, MR-Eagle method and weighted median method. The Cochrane Q-test, MR-Egger interception, MR-multidirectional residual and outlier methods, entrance/exit analysis and funnel plot were applied for sensitivity analysis. Two-sample Mendelian randomization analysis of the 19 SNPs obtained from screening showed no significant causal effect of SLE and osteonecrosis, and IVW and MR-Egger heterogeneity analyses showed no significant heterogeneity between the instrumental variables (P > 0.05). Multi-phenotype MR analysis showed no significant causal effect between gout and rheumatoid arthritis and osteonecrosis (p > 0.05). The available evidence does not support a significant causal effect of gout and rheumatoid arthritis on osteonecrosis, and the causal effect of SLE on the increased risk of osteonecrosis is only supported by the IVW method, which is of insufficient evidence validity, but suggests a better theoretical basis for the study of heritability related to SLE.
Collapse
Affiliation(s)
- Baochuang Qi
- First Clinical Medical College of Yunnan University of Chinese Medicine / Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, 650500, Yunnan, China
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Jinglin Zhao
- Medical Laboratory, Kunming Children's Hospital, Kunming, 650000, Yunnan, China
| | - Chen Meng
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Pengfei Bu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Lu Qiao Pu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wanqiu Zhao
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Chuan Li
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China.
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Yongqing Xu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China.
| |
Collapse
|
10
|
Zhang J, Zhuang W, Li Y, Deng C, Xuan J, Sun Y, He Y. Bioinformatic analysis and experimental verification reveal expansion of monocyte subsets with an interferon signature in systemic lupus erythematosus patients. Arthritis Res Ther 2025; 27:96. [PMID: 40281593 PMCID: PMC12023529 DOI: 10.1186/s13075-025-03560-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by chronic inflammation and multi-organ damage. A central factor in SLE pathogenesis is the excessive production of type I interferon (IFN-I), which drives immune dysregulation. Monocytes, key components of the immune system, significantly contribute to IFN-I production. However, their specific roles in SLE remain incompletely understood. METHODS This study utilized bioinformatics and statistical analyses, including robust rank aggregation (RRA), DESeq2, and limma, to analyze transcriptome data from peripheral blood mononuclear cells (PBMCs) and monocytes of SLE patients and healthy controls. Single-cell RNA sequencing (scRNA-seq) data were processed using the Seurat R package to identify and characterize monocyte subsets with a strong IFN-driven gene signature. Flow cytometry was employed to validate key findings, using markers such as CD14, SIGLEC1, and IRF7 to confirm monocyte subset composition. RESULTS Our research has found that monocytes in SLE undergo IFN-driven transcriptional reprogramming, with the upregulation of key interferon signature genes (ISGs), forming the SLE-Related Monocyte Signature (SLERRAsignature). Moreover, the composition of mononuclear phagocyte subsets in SLE patients changes, with an increase trend in the proportion of the CD14Mono8 subset in the flare group. The differentially expressed genes (DEGs) in 13 mononuclear phagocyte subsets of SLE are mainly ISGs, and the expression of ISGs is higher in severe patients. We identified SIGLEC1+IRF7+ monocytes among these subsets and for the first time discovered this group of cells in the peripheral blood of healthy individuals. In SLE, the enrichment score of the gene set representing SIGLEC1+IRF7+ monocytes is positively correlated with the severity of SLE. Finally, flow cytometry confirmed that the frequency of CD14+SIGLEC1+IRF7+ monocytes in PBMCs was higher in SLE compared with healthy controls. CONCLUSIONS Our study found that the expansion of IFN-I-producing monocyte subsets, particularly the CD14+SIGLEC1+IRF7+ subset, plays a crucial role in SLE pathogenesis. This subset may serve as a potential biomarker and therapeutic target for managing SLE.
Collapse
Affiliation(s)
- Jimin Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China
| | - Wuwei Zhuang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China
| | - Yan Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China
| | - Chaoqiong Deng
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China
| | - Jingxiu Xuan
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China
| | - Yuechi Sun
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China.
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China.
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China.
| | - Yan He
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, XM, China.
- Xiamen Municipal Clinical Research Center for Immune Diseases, Xiamen, 361000, XM, China.
- Xiamen Key Laboratory of Rheumatology and Clinical Immunology, Xiamen, 361000, XM, China.
| |
Collapse
|
11
|
Zeng Z, Mao H, Lei Q, He Y. IL-7 in autoimmune diseases: mechanisms and therapeutic potential. Front Immunol 2025; 16:1545760. [PMID: 40313966 PMCID: PMC12043607 DOI: 10.3389/fimmu.2025.1545760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/14/2025] [Indexed: 05/03/2025] Open
Abstract
Interleukin-7 (IL-7) is a pleiotropic cytokine that plays a crucial role in the development, homeostasis, and function of the immune system. Growing evidence has demonstrated that IL-7 is involved in the pathogenesis of various autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D) and multiple sclerosis (MS). This review aims to summarize the current understanding of the role of IL-7 in autoimmune diseases, focusing on its mechanisms of action, implications for disease progression, and potential therapeutic applications. Produced by stromal cells, IL-7 binds to IL-7 receptor (IL-7R) on diverse immune cells. It is crucial for T cell development, survival, and proliferation. In autoimmune diseases, it activates and expands autoreactive T cells and influences B cell function, potentially leading to autoantibody production. The review further delves into the role of IL-7 in different autoimmune diseases. In RA, elevated IL-7/IL-7R promotes memory T cell survival, cytokine production, and influences B cells and monocytes to contribute to inflammation and joint damage. In SLE, elevated soluble form of IL-7R is associated with disease activity, promoting the survival of autoreactive T cells and enhancing the production of pro-inflammatory cytokines. In MS, genetic variations in the IL-7R gene are linked to disease susceptibility, and IL-7 impacts the survival and differentiation of T cell subsets involved in multiple sclerosis pathogenesis. For T1D, IL-7 affects the function of immune cells that attack pancreatic β cells. Given its central role in autoimmune processes, targeting the IL-7/IL-7R axis holds great therapeutic potential. By modulating IL-7 signaling, it may be possible to restore immune tolerance, reduce the activation of autoreactive immune cells, and alleviate disease symptoms. Understanding the complex mechanisms of IL-7 in autoimmune diseases is essential for the development of effective and targeted therapies.
Collapse
Affiliation(s)
| | | | | | - Yuanmin He
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
12
|
Richter P, Macovei LA, Rezus C, Boiculese VL, Buliga-Finis ON, Rezus E. IL-10 in Systemic Lupus Erythematosus: Balancing Immunoregulation and Autoimmunity. Int J Mol Sci 2025; 26:3290. [PMID: 40244128 PMCID: PMC11989541 DOI: 10.3390/ijms26073290] [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: 03/07/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Interleukin-10 (IL-10) presents a dual role in systemic lupus erythematosus (SLE), illustrating pro-inflammatory and anti-inflammatory effects. This study addressed the possible associations between serum IL-10 levels and demographic characteristics, laboratory parameters, organ manifestations, disease activity, and treatment response in SLE patients. A total of 88 SLE patients from the Rheumatology Clinic of the Clinical Rehabilitation Hospital, Iași, were enrolled. Disease activity was assessed using the Systemic Lupus Erythematosus Disease Activity Index (SLEDAI). Serum cytokine levels were measured using an enzyme-linked immunosorbent assay (ELISA). Serum IL-10 levels were significantly higher in males compared to females (47.62 pg/mL vs. 13.24 pg/mL, p = 0.011) but not significantly associated with age or disease duration. However, IL-10 showed positive correlations with inflammatory markers and autoantibodies, including C-reactive protein (p = 0.002), IL-6 (p = 0.01), ANA (p = 0.014), and anti-SSB antibodies (p = 0.005). Our findings indicate that IL-10 may be involved in inflammatory and immune processes in SLE, as evidenced by its significant correlations with specific autoantibodies and inflammatory markers in our study. However, IL-10 did not correlate with disease activity, organ involvement, or treatment response. These results underline the participation of IL-10 in SLE and emphasize the need for further research to clarify its potential as a biomarker or therapeutic target.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (P.R.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (P.R.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Vasile Lucian Boiculese
- Department of Medical Informatics and Biostatistics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Oana Nicoleta Buliga-Finis
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
| | - Elena Rezus
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (P.R.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| |
Collapse
|
13
|
Gupta D, Palayullakandi A, Sopanam S, Panda PK, Sharawat IK. Opsoclonus Myoclonus Ataxia Syndrome: An Atypical Presentation of Tuberculous Meningitis. Am J Trop Med Hyg 2025; 112:856-858. [PMID: 39773430 PMCID: PMC11965770 DOI: 10.4269/ajtmh.24-0664] [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: 10/02/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025] Open
Abstract
Opsoclonus myoclonus ataxia syndrome (OMAS) is a rare neuroinflammatory disorder that is typically associated with paraneoplastic and postinfectious processes. Opsoclonus myoclonus ataxia syndrome has not been previously reported in association with tuberculous meningitis (TBM). This report presents a unique case in which TBM manifested as OMAS, highlighting the complex interplay between tuberculosis and autoimmune neurological conditions. A 1.5-year-old previously healthy girl, presented with acute-onset jerky movements, opsoclonus, irritability, and reduced sleep over 4 weeks. A neurological examination revealed opsoclonus, irritability, generalized tremulousness, and fragmentary myoclonus. Neuroimaging was suggestive of TBM. A cerebrospinal fluid (CSF) analysis indicated lymphocytic pleocytosis with positive CSF cartridge-based nucleic acid amplification test results for tuberculosis. The patient was treated with methylprednisolone pulse therapy, intravenous immunoglobulins, and anti-tuberculous therapy (ATT). Significant symptom improvement was observed within 2 weeks. This case underscores a rare association between OMAS and TBM, demonstrating that tuberculosis can trigger OMAS through autoimmune mechanisms. A timely diagnosis and treatment with ATT and immunotherapy can lead to substantial recovery.
Collapse
Affiliation(s)
- Diksha Gupta
- Pediatric Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, India
| | - Achanya Palayullakandi
- Pediatric Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, India
| | - Suthiraj Sopanam
- Pediatric Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, India
| | - Prateek Kumar Panda
- Pediatric Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, India
| | - Indar Kumar Sharawat
- Pediatric Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, India
| |
Collapse
|
14
|
Hu Y, Huang J, Wang S, Sun X, Wang X, Yu H. Deciphering Autoimmune Diseases: Unveiling the Diagnostic, Therapeutic, and Prognostic Potential of Immune Repertoire Sequencing. Inflammation 2025; 48:676-695. [PMID: 38914737 DOI: 10.1007/s10753-024-02079-2] [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: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Autoimmune diseases (AIDs) are immune system disorders where the body exhibits an immune response to its own antigens, causing damage to its own tissues and organs. The pathogenesis of AIDs is incompletely understood. However, recent advances in immune repertoire sequencing (IR-seq) technology have opened-up a new avenue to study the IR. These studies have revealed the prevalence in IR alterations, potentially inducing AIDs by disrupting immune tolerance and thereby contributing to our comprehension of AIDs. IR-seq harbors significant potential for the clinical diagnosis, personalized treatment, and prognosis of AIDs. This article reviews the application and progress of IR-seq in diseases, such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes, to enhance our understanding of the pathogenesis of AIDs and offer valuable references for the diagnosis and treatment of AIDs.
Collapse
Affiliation(s)
- Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China.
| |
Collapse
|
15
|
Hong Y, Wang D, Qian H, Jiang X, Wang Y, Liang X, Gao S, Hua C. Exploring the molecular mechanism of Tripterygium Wilfordii Hook F in treating systemic lupus erythematosus via network pharmacology and molecular docking. Clin Rheumatol 2025; 44:1549-1569. [PMID: 39934490 DOI: 10.1007/s10067-025-07311-4] [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: 05/11/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Tripterygium wilfordii Hook F (TwHF) is a prominent Chinese herbal formula. It exhibits significant clinical efficacy in treating systemic lupus erythematosus (SLE), though its mechanisms remain unclear. Our study employs network pharmacology and molecular docking to explore active compounds of TwHF and their associated targets for SLE treatment. METHODS Primary active compounds of TwHF and their targets were sourced from the TCMSP, SwissTargetPrediction, and UniProt databases. SLE-relevant target proteins were identified from the OMIM and GeneCards databases. Enrichment analyses were conducted to reveal results of common TwHF-SLE targets. STRING and Cytoscape software were used to systematically analyze and construct protein-protein interaction (PPI) networks, compound-target-pathway, and target-organ networks. Molecular docking was utilized to confirm the binding of key targets to the top active compounds. RESULTS A total of 14 active compounds and 300 overlapping targets between TwHF and SLE were identified. PPI network analysis revealed 29 core targets. Several pathways were found to contribute to the potential therapeutic effects of TwHF in SLE, including PI3K-Akt signaling pathway, Th17 cell differentiation, chemokine signaling, and T cell receptor signaling. Disease Ontology (DO) analysis highlighted the involvement of TwHF in genes associated with myocardial infarction (MI), atherosclerosis (AS), breast carcinoma, and ischemia. Molecular docking results demonstrated strong binding affinities, with 37 signal molecule-receptor interactions in SLE and 97 interactions in SLE-related MI and AS showing binding energies lower than -7 kJ/mol. CONCLUSIONS This research effectively anticipates the potent constituents, probable targets, and pathways implicated in treating SLE with TwHF, specifically addressing complications such as MI and AS. Comprehending the precise molecular mechanism targeting SLE of TwHF and its efficacious bioactive components furnishes a theoretical groundwork for enhancing its clinical utilization. Key Points •SLE is characterized by aberrant immune activation and persistent inflammation. •TwHF exerts immunomodulatory and anti-inflammatory effects. •TwHF exhibits prospects in the treatment of SLE with unknown molecular mechanisms. •Network pharmacology and molecular docking reveal promise in the mechanism of TwHF.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Deqi Wang
- The First School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Hengrong Qian
- The Second School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Xiaoyang Jiang
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Yi Wang
- The First School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Xinyue Liang
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| |
Collapse
|
16
|
Priya K, Rawat S, Das D, Chaubey M, Thacker H, Giri K, Singh S, Rai M, Mohanty S, Rai G. Autoimmunity and clinical pathology amelioration in SLE by dexamethasone primed mesenchymal stem cell derived conditioned media. Stem Cell Res Ther 2025; 16:158. [PMID: 40158179 PMCID: PMC11954324 DOI: 10.1186/s13287-025-04208-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/29/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND This study aimed to investigate the therapeutic potential of cell-free Dexamethasone (Dex) primed Wharton's jelly Mesenchymal stem cells derived conditioned media (DW) in addressing complications associated with systemic lupus erythematosus (SLE), focusing on its immunomodulatory effects. METHODS Peripheral blood mononuclear cells from 74 SLE patients were stimulated and treated with Dex, DW and W. Culture supernatant were evaluated for autoantibody levels, IL-10 and TGF-β by ELISA, Treg subtypes, Breg subtypes, TH17 cells Double negative T cells and inflammatory neutrophils by flow cytometry, IL-10 and IL-17A by qPCR. In vivo studies were performed on 60 pristane induced female BALB/c mice. Dex and DW treatments were evaluated for autoantibody production, proteinuria, immunomodulation of immune cells, organ function, and histopathology. In vivo imaging of internal organs was done using VevoLAZR-X photoacoustic imaging system. RESULTS DW treatment significantly expanded different Treg and Bregs subtypes. DW suppressed pathogenic TH17, Double negative T cells and inflammatory neutrophils. Comparative analyses with hydroxychloroquine showed similar effects, with combined treatment enhancing efficacy. Inhibition studies implicated the TGF-β pathway in DW's mechanism. In vivo studies using the PIL mouse model showed that DW treatment reduced mortality, prevented proteinuria, and ameliorated symptoms such as limb inflammation, seizures, and alopecia. Detailed organ-specific evaluations through live imaging and histopathological analyses revealed DW's protective effects on kidneys, liver, lungs, heart, and spleen. CONCLUSION DW shows promise as a cell-free biological therapy for SLE and related autoimmune disorders, capable of modulating immune responses effectively without the adverse effects of glucocorticoids.
Collapse
Affiliation(s)
- Khushbu Priya
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Sonali Rawat
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, 110029, India
| | - Doli Das
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Manaswi Chaubey
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Hiral Thacker
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Kiran Giri
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | | | - Madhukar Rai
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, 110029, India
| | - Geeta Rai
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
17
|
Cui X, Cong Y. Role of Gut Microbiota in the Development of Some Autoimmune Diseases. J Inflamm Res 2025; 18:4409-4419. [PMID: 40162082 PMCID: PMC11954480 DOI: 10.2147/jir.s515618] [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: 01/03/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiota is crucial for maintaining the homeostasis and function of the immune system. It interacts with the host's immune system through various mechanisms, including promoting immune tolerance, affecting the differentiation and function of immune cells, and participating in the metabolism of immune regulatory substances. The disruption of the gut microbiome may lead to impaired mucosal barrier function, allowing bacteria and their metabolites to invade into the host, activate or interfere with the immune system, and potentially trigger or exacerbate autoimmune responses. Understanding the relationship between the microbiome and autoimmune diseases may help develop new treatment strategies. This article reviewed the recent progresses of microbiome involved in the occurrence and development of some autoimmune diseases and the treatment methods based on regulation of the microbiome, highlighted the key role of microbiome in autoimmune diseases.
Collapse
Affiliation(s)
- Xiaojing Cui
- Department of Clinical Laboratory, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong Province, 523710, People’s Republic of China
- Dongguan Key Laboratory for Pathogenesis and Experimental Diagnosis of Infectious Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong Province, 523710, People’s Republic of China
| | - Yanguang Cong
- Department of Clinical Laboratory, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong Province, 523710, People’s Republic of China
- Dongguan Key Laboratory for Pathogenesis and Experimental Diagnosis of Infectious Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong Province, 523710, People’s Republic of China
| |
Collapse
|
18
|
Cho DY, Kang YM, Seol S. Suppression of skin lesions and SLE nephritis by increasing Treg in MRL/FAS lpr mice by administration of bee venom Apitoxin ®. Adv Rheumatol 2025; 65:17. [PMID: 40108667 DOI: 10.1186/s42358-025-00448-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Apitoxin®, a drug based on bee venom was approved and released in Korea in 2003 as the ethical drug (ETC). It is well-known for its pain-relieving properties due to its potent anti-inflammatory effects. This raises the question of whether bee venom has benefits for other inflammatory disorders. Since its effectiveness in treating inflammation and pain associated with autoimmune diseases has been observed in several clinical cases in Korea, we conducted an efficacy study using an animal model of the systemic lupus erythematosus (SLE), an autoimmune disease with high medical unmet needs. In this research, we aim to confirm the potential therapeutic efficacy for SLE through the immunomodulation induced by bee venom. METHODS MRL/FASlpr mice were injected subcutaneously with Apitoxin® and evaluated for clinical parameters including proteinuria, skin lesions, and lymphadenopathy, flow cytometric evaluation of regulatory T cells (Treg), quantitative evaluation of anti-dsDNA antibody in serum by ELISA, and histomorphometric analysis of kidney tissues. RESULTS Treatment with Apitoxin® revealed a reduction in proteinuria, skin lesions, and lymphadenopathy in MRL/FASlpr mice. The percentage of CD3+CD4+CD25+FoxP3 (Treg) cells, which are associated with autoimmune diseases, was increased compared to the negative control (vehicle). Quantitative analysis of autoantibodies in the blood of MRL/FASlpr mice showed a decreasing tendency in the treatment groups with Apitoxin®. Moreover, mesangial proliferation and inflammatory cell infiltration in glomeruli were significantly reduced in the treatment group with Apitoxin®, which was associated with a statistically significant decrease in the amount of IgG infiltrated into the glomeruli. CONCLUSION Overall, the results confirmed that Apitoxin® induced clinical improvement in SLE by increasing the proportion of Treg cells and decreasing anti-dsDNA antibodies in the blood, which resulted in therapeutic effects on glomerulonephritis associated with decreased renal infiltration of immune complexes.
Collapse
Affiliation(s)
- Duk-Yeon Cho
- APIMEDS, Inc., 608, Namsung Plaza, 130, Digital-ro, Geumcheon-gu, Seoul, Republic of Korea
| | - Young-Mo Kang
- Preclina Inc., 719, Tera Tower B, 167, Songpa-daero, Songpa-gu, Seoul, Republic of Korea
| | - SangHo Seol
- APIMEDS, Inc., 608, Namsung Plaza, 130, Digital-ro, Geumcheon-gu, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Royo M, Joseph-Mullol B, Sandoval S, Moliné T, Solé C, Cortés-Hernández J. Integrative miRNA-mRNA profiling uncovers mechanisms of belimumab action in systemic lupus erythematosus. Front Immunol 2025; 16:1553971. [PMID: 40160819 PMCID: PMC11949941 DOI: 10.3389/fimmu.2025.1553971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder driven by autoreactive B cells and characterized by the production of pathogenic autoantibodies. Belimumab, an anti-BAFF monoclonal antibody, has demonstrated efficacy in reducing disease activity and corticosteroid use in SLE patients, although responses remain variable. B-cell activating factor (BAFF) is essential for B cell survival and autoantibody production, positioning it as a key target in SLE pathogenesis. MicroRNAs (miRNAs), critical regulators of gene expression and immune homeostasis, have an emerging role in SLE pathophysiology. However, their regulation in response to anti-BAFF therapies, such as belimumab, remains unexplored. This study investigates miRNA-mRNA interactions in T cells, B cells, and myeloid cells from SLE patients before and after belimumab treatment. A total of 79 miRNAs associated with treatment response and 525 miRNA-gene interactions were identified. Validation in 18 SLE responders revealed significant changes in miRNA expression in T and myeloid cells, but not in B cells. Belimumab was found to modulate B cell development by regulating genes such as BLNK, BANK1, and MEF2C, as well as the CD40/CD40L axis. In T cells, miRNAs influenced interferon signaling and inflammatory cytokines via NF-κB activation. Changes in myeloid cells, characterized by the downregulation of KLF13, CCL5, and IL4, appear to be secondary to T cell modulation. These findings provide novel insights into the miRNA-mediated regulatory networks underlying belimumab's immunomodulatory effects in SLE. Further research is required to validate these findings and through in vitro experiments to better understand the role of miRNAs in guiding treatment responses.
Collapse
Affiliation(s)
- Maria Royo
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Blanca Joseph-Mullol
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sebastian Sandoval
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa Moliné
- Department of Pathology, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Solé
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Josefina Cortés-Hernández
- Rheumatology Research Group, Lupus Unit, Hospital Universitari Vall d’Hebron, Institut de Recerca (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Yuan Z, Liu M, Zhang L, Jia L, Hao S, Su D, Tang L, Wang C, Wang M, Wen Z. Notch1 hyperactivity drives ubiquitination of NOX2 and dysfunction of CD8+ regulatory T cells in patients with systemic lupus erythematosus. Rheumatology (Oxford) 2025; 64:1500-1512. [PMID: 38652598 DOI: 10.1093/rheumatology/keae231] [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: 01/11/2024] [Revised: 03/24/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVES Patients with SLE display heightened immune activation and elevated IgG autoantibody levels, indicating compromised regulatory T cell (Tregs) function. Our recent findings pinpoint CD8+ Tregs as crucial regulators within secondary lymphoid organs, operating in a NOX2-dependent mechanism. However, the specific involvement of CD8+ Tregs in SLE pathogenesis and the mechanisms underlying their role remain uncertain. METHODS SLE and healthy individuals were enlisted to assess the quantity and efficacy of Tregs. CD8+CD45RA+CCR7+ Tregs were generated ex vivo, and their suppressive capability was gauged by measuring pZAP70 levels in targeted T cells. Notch1 activity was evaluated by examining activated Notch1 and HES1, with manipulation of Notch1 accomplished with Notch inhibitor DAPT, Notch1 shRNA, and Notch1-ICD. To create humanized SLE chimaeras, immune-deficient NSG mice were engrafted with PBMCs from SLE patients. RESULTS We observed a reduced frequency and impaired functionality of CD8+ Tregs in SLE patients. There was a downregulation of NOX2 in CD8+ Tregs from SLE patients, leading to a dysfunction. Mechanistically, the reduction of NOX2 in SLE CD8+ Tregs occurred at a post-translational level rather than at the transcriptional level. SLE CD8+ Tregs exhibited heightened Notch1 activity, resulting in increased expression of STUB1, an E3 ubiquitin ligase that binds to NOX2 and facilitates its ubiquitination. Consequently, restoring NOX2 levels and inhibiting Notch1 activity could alleviate the severity of the disease in humanized SLE chimaeras. CONCLUSION Notch1 is the cell-intrinsic mechanism underlying NOX2 deficiency and CD8+ Treg dysfunction, serving as a therapeutic target for the clinical management of SLE.
Collapse
Affiliation(s)
- Zixin Yuan
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Mengdi Liu
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Li Jia
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Siao Hao
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Danhua Su
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| | - Longhai Tang
- Division of Research Center, Suzhou Blood Center, Suzhou, China
| | - Chunhong Wang
- Cyrus Tang Hematology Center, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Mingyuan Wang
- Division of Research Center, Suzhou Blood Center, Suzhou, China
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China
| |
Collapse
|
21
|
Wójcicka-Frankiewicz A, Pawlak MA, Piernik M, Forycka M, Wiłkość-Dębczyńska M, Więsik-Szewczyk E, Pawlak-Buś K, Leszczyński P, Michalak S, Wypych A, Serafin Z, Kozubski W, Kalinowska-Łyszczarz A. The impact of white matter brain lesions on processing speed deficits in systemic lupus erythematosus patients. Lupus 2025; 34:270-280. [PMID: 39927750 DOI: 10.1177/09612033251319826] [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: 02/11/2025]
Abstract
BackgroundThe mechanism of cognitive dysfunction in systemic lupus erythematosus (SLE) is still not fully understood. Even though many SLE patients present some neurological dysfunction, including various cognitive deficits, neither a specific pattern of cognitive dysfunction nor specific structural changes associated with cognitive impairment in SLE patients have been established. Moreover, although prevalent and bothersome, cognitive deficits have not been included in the most recent SLE diagnostic criteria.PurposeThe aim of this study was to determine the relationship between the presence of white matter lesions (WMLs) and cognitive dysfunction in patients diagnosed with SLE.Research Design33 SLE patients underwent 3 T brain magnetic resonance imagining (MRI) and an extensive battery of psychological tests, including Automated Neuropsychological Assessment Metrics (ANAM) and the standard pen and paper neuropsychological tests. Patients were stratified into two groups based on the presence (N = 15) or absence (N = 18) of WMLs. Psychometric scores were compared between the two groups.Results and conclusionsSignificant deficits in cognitive functions were observed. Patients with WMLs showed deficits in attention and executive functions, as well as memory deficits in comparison to the group without WMLs. As measured with: STROOP Test (executive function), Color Trail Test (CTT) (attention), Californian Verbal Learning Test (CVLT) (memory), and from ANAM tests with: Procedural Reaction Time (PRT) (attention), Code Substitution Delayed (CS_D) (memory), Spatial Processing (SP) (visuospatial functions), Tower Puzzle (TP) (executive functions), 2 Choice Reaction Time (2CHRT) (attention), Running Memory CPT (RM CPT) (memory), Matching Grids (Mat GR) (visuospatial functions), Go/No _Go inhibition (Go/No_Go Go) (executive functions). Additionally, we analyzed structural volumetric measures derived from a comprehensive segmentation pipeline recon-all using Freesurfer 5.3. Significant differences were identified for the following structures' volumes: right choroid plexus, left choroid plexus, right lateral ventricle. All these structures had a greater volume in patients with WMLs.
Collapse
Affiliation(s)
| | - Mikołaj A Pawlak
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Piernik
- Institute of Computing Science, Poznan University of Technology, Poznan, Poland
| | - Maria Forycka
- Chair of Palliative Medicine, Institute of Medical Sciences Collegium Medicum, University of Zielona Góra, Zielona Góra, Poland
| | - Monika Wiłkość-Dębczyńska
- Department of Health Psychology, Faculty of Psychology, Kazimierz Wielki University in Bydgoszcz, Bydgoszcz, Poland
| | - Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergy, Clinical Immunology and Rare Diseases, Central Clinical Hospital of the Ministry of National Defence, Military Institute of Medicine, National Science Institute, Warsaw, Poland
| | - Katarzyna Pawlak-Buś
- Department of Internal Medicine, Poznan University of Medical Sciences, Poznań, Poland
- Department of Rheumatology, Systemic Connective Tissue Diseases and Immunotherapy of Rheumatic Diseases, J. Struś Municipal Hospital, Poznań, Poland
| | - Piotr Leszczyński
- Department of Internal Medicine, Poznan University of Medical Sciences, Poznań, Poland
- Department of Rheumatology, Systemic Connective Tissue Diseases and Immunotherapy of Rheumatic Diseases, J. Struś Municipal Hospital, Poznań, Poland
| | - Sławomir Michalak
- Division of Neurochemistry and Neuropathology, Department of Neurology, Department of Neurosurgery and Neurotraumatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Aleksandra Wypych
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
| | - Zbigniew Serafin
- Department of Radiology and Diagnostic Imaging, Nicolaus Copernicus University, Collegium Medicum, Bydgoszcz, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Alicja Kalinowska-Łyszczarz
- Division of Neurochemistry and Neuropathology, Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
22
|
Dudkova M, Petrackova A, Radvansky M, Skacelova M, Videman J, Manakova J, Kraiczova VS, Kudelka M, Smrzova A, Langova K, Mrazek F, Kriegova E, Horak P. Blood gene expression of Toll-like receptors in SLE patients with lupus nephritis or neuropsychiatric systemic lupus erythematosus. Arthritis Res Ther 2025; 27:41. [PMID: 40011954 PMCID: PMC11863772 DOI: 10.1186/s13075-025-03512-z] [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: 06/25/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND To determine differences in the blood innate gene expression signatures of systemic lupus erythematosus (SLE) patients across various organ manifestations and disease activity, with a focus on lupus nephritis (LN) and central nervous system (CNS) involvement. METHODS Toll-like receptor family (TLR 1-10) mRNA expression was investigated in peripheral blood mononuclear cells from patients with SLE (n = 74) and healthy controls (n = 34). We compared patients with histologically confirmed active LN or neuropsychiatric systemic lupus erythematosus (NPSLE) with patients without these symptoms. The expression of TLR mRNA was determined by RT‒qPCR using a high-throughput SmartChip Real-Time-qPCR system (WaferGen). Multivariate analysis and nonparametric statistics were used for data analysis to assess the associations between TLRs and disease activity and severity. RESULTS TLR4 (0.044 vs. 0.081, p = 0.012) was upregulated and TLR10 (0.009 vs. 0.006, p = 0.0007) was downregulated in the whole cohort of SLE patients compared to healthy controls. A comparison of the active LN group with participants without kidney involvement revealed increased expression of TLR2 (0.078 vs. 0.03, p = 0.009), and TLR5 (0.035 vs. 0.017, p = 0.03). Moreover, a significant difference was observed in TLR9 expression between inactive LN and the control group (0.014 vs. 0.009, p = 0.01), together with borderline correlation in TLR2 expression (0.04 vs. 0.03, p = 0.06). Receiver operating characteristic (ROC) curve analysis revealed that TLR1 and TLR2 expression were the best potential diagnostic markers for active LN. The NPSLE group showed upregulation of TLR1 (0.088 vs. 0.048, p = 0.01), TLR4 (0.173 vs. 0.066, p = 0.0003) and TLR6 (0.087 vs. 0.036, 0.007). Our correlation analysis supported the close relationships among the expression of individual TLRs in the whole lupus cohort and its subgroups. CONCLUSION Our study revealed differences in TLR expression between a lupus cohort and healthy controls. Additionally, our analysis provides insight into specific TLR expression in cases with severe organ manifestations, such as LN and NPSLE. The multiple mutual relationships of TLRs demonstrate the activation of innate immunity in SLE and suggest promising targets for future therapies or diagnostics.
Collapse
Affiliation(s)
- Marketa Dudkova
- Department of Internal Medicine III - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Anna Petrackova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Martin Radvansky
- Department of Computer Science, Faculty of Electrical Engineering and Computer Science, Technical University of Ostrava, Ostrava, Czech Republic
| | - Martina Skacelova
- Department of Internal Medicine III - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Jakub Videman
- Department of Internal Medicine III - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Jirina Manakova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Veronika Smotkova Kraiczova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Milos Kudelka
- Department of Computer Science, Faculty of Electrical Engineering and Computer Science, Technical University of Ostrava, Ostrava, Czech Republic
| | - Andrea Smrzova
- Department of Internal Medicine III - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Katerina Langova
- Department of Medical Biophysics, Czech Republic, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Frantisek Mrazek
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Pavel Horak
- Department of Internal Medicine III - Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic.
- III. Interni Klinika - Nefrologicka, Revmatologicka a Endokrinologicka, Fakultni Nemocnice Olomouc, Zdravotniku 248/7, Olomouc, 779 00, Czech Republic.
| |
Collapse
|
23
|
Giri S, Batra L. Memory Cells in Infection and Autoimmunity: Mechanisms, Functions, and Therapeutic Implications. Vaccines (Basel) 2025; 13:205. [PMID: 40006751 PMCID: PMC11860616 DOI: 10.3390/vaccines13020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Memory cells are central to the adaptive immune system's ability to remember and respond effectively to previously encountered pathogens. While memory cells provide robust protection against infections, they can also contribute to autoimmunity when regulation fails. Here, we review the roles of memory T and B cells in infection and autoimmunity, focusing on their differentiation, activation, effector functions, and underlying regulatory mechanisms. We elaborate on the precise mechanisms by which memory cells contribute to autoimmune diseases, highlighting insights from current research on how pathogenic memory responses are formed and sustained in autoimmunity. Finally, we explore potential therapeutic strategies aimed at modulating memory cells to prevent or treat autoimmune disorders, including B cell-depleting therapies (e.g., Rituximab), T cell-targeting agents (e.g., Abatacept), and cytokine inhibitors (e.g., IL-17 or IL-23 blockers) that are currently used in diseases such as rheumatoid arthritis, multiple sclerosis, and psoriasis.
Collapse
Affiliation(s)
- Shilpi Giri
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lalit Batra
- Center for Predictive Medicine for Biodefence and Emerging Infectious Diseases, School of Medicine, University of Louisville, Louisville, KY 40222, USA;
| |
Collapse
|
24
|
Predescu OR, Vreju FA, Musetescu AE, Florescu A, Dinescu SC, Bita CE, Barbulescu AL, Ciurea PL. Effects of Vitamin D Supplementation on Fatigue and Disease Activity in Systemic Lupus Erythematosus. Cureus 2025; 17:e78830. [PMID: 40084313 PMCID: PMC11906197 DOI: 10.7759/cureus.78830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by chronic inflammation and various clinical symptoms, with vitamin D deficiency suggested as a contributing factor. This study aimed to evaluate the effects of vitamin D supplementation on fatigue and disease activity in SLE patients. METHODS Patients diagnosed based on EULAR/ACR 2019 criteria were divided into three groups: no supplementation, 4000 IU, and 8000 IU of vitamin D daily for six months. Clinical assessments included serum complement levels (C3 and C4), fatigue scores (Functional Assessment of Chronic Illness Therapy (FACIT)-Fatigue and Fatigue Severity Scale (FSS)), and disease activity (Safety of Estrogens in Lupus Erythematosus National Assessment-Systemic Lupus Erythematosus Disease Activity Index (SELENA-SLEDAI)). RESULTS Results showed significant increases in vitamin D levels and serum complement levels in the supplementation groups. Serum complement levels and fatigue scores improved significantly in both the 4000 IU and 8000 IU groups. Additionally, there was a slight reduction in SELENA-SLEDAI scores in the treated groups, but without statistical significance. CONCLUSIONS The findings suggest that vitamin D supplementation positively affects fatigue and some parameters of disease activity in SLE patients, though its overall impact on disease activity needs further investigation.
Collapse
Affiliation(s)
| | - Florentin Ananu Vreju
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | | | - Alesandra Florescu
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | | | - Cristina Elena Bita
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| | | | - Paulina Lucia Ciurea
- Department of Rheumatology, University of Medicine and Pharmacy of Craiova, Craiova, ROU
| |
Collapse
|
25
|
Zhu BY, Liu ZC, Zhao ZX, Huang HP, Zhang N, Xia J, Chen WW. Pharmacological Mechanism of Chinese Medicine in Systemic Lupus Erythematosus: A Narrative Review. Chin J Integr Med 2025; 31:157-169. [PMID: 39240290 DOI: 10.1007/s11655-024-3762-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2024] [Indexed: 09/07/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder affecting multiple systems, characterized by the development of harmful autoantibodies and immune complexes that lead to damage in organs and tissues. Chinese medicine (CM) plays a role in mitigating complications, enhancing treatment effectiveness, and reducing toxicity of concurrent medications, and ensuring a safe pregnancy. However, CM mainly solves the disease comprehensively through multi-target and multi-channel regulation process, therefore, its treatment mechanism is often complicated, involving many molecular links. This review introduces the research progress of pathogenesis of SLE from the aspects of genetics, epigenetics, innate immunity and acquired immunity, and then discusses the molecular mechanism and target of single Chinese herbal medicine and prescription that are commonly used and effective in clinic to treat SLE.
Collapse
Affiliation(s)
- Bo-Yu Zhu
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhi-Chao Liu
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Zhen-Xi Zhao
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Hui-Ping Huang
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Na Zhang
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Jia Xia
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Wei-Wei Chen
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
26
|
Philip R, Elhani I, Gallou S, Boysson HD, Martin Silva N, Georgin-Lavialle S, Deshayes S, Aouba A. A20 haploinsufficiency diagnosis beyond systemic lupus erythematosus: A systematic review of the literature. Autoimmun Rev 2025; 24:103722. [PMID: 39672252 DOI: 10.1016/j.autrev.2024.103722] [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: 09/18/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease whose pathophysiology remains incompletely understood, involving genetic and epigenetic factors. However, an increasing small subset of patients present with monogenic lupus, providing insight into the pathogenesis of the disease. This systematic review focuses on SLE associated with A20 haploinsufficiency (HA20), a monogenic disorder associated with tumor necrosis factor alpha-induced protein 3 gene (TNFAIP3) variants. Besides the mainly auto-inflammatory phenotypic expression of HA20 mimicking Behçet's disease spectrum, some of its clinical and biological manifestations are part of the spectrum of autoimmune diseases, including glomerulonephritis as well as the frequent presence of antinuclear antibodies, sometimes with anti-DNA specificity. Among all the 191 HA20 patients reported in the literature, we identified 16 patients (8.4 %) with a compatible diagnosis of SLE. When estimable, the SLICC 2012 and EULAR/ACR 2019 classification criteria were positive for 92.9 % of them. A majority had multi-system involvement, mainly cutaneous (81.3 %), musculoskeletal (56.3 %), and/or renal (56.3 %) manifestations. They also seemed to exhibit differences compared to other SLE patients: higher prevalence of fever, chronic cutaneous lupus erythematosus, oral and genital ulcers, neuropsychiatric manifestations, autoimmune cytopenia, and elevated biologic inflammatory markers. This review highlights the necessity of considering TNFAIP3 variants in SLE patients with early-onset disease, familial history, and/or specific clinical manifestations suggestive of autoinflammatory diseases. Recognizing HA20-SLE patients may improve our understanding of SLE pathogenesis and lead to better therapeutic strategies for these patients.
Collapse
Affiliation(s)
- Rémi Philip
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France.
| | - Inès Elhani
- AP-HP, Tenon Hospital, Department of Internal Medicine, Paris, France; Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA) INSERM UMRS-938, USA; National French Reference Centre for Auto-inflammatory Diseases and Inflammatory Amyloidosis (CEREMAIA), France
| | - Sophie Gallou
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| | - Hubert De Boysson
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| | - Nicolas Martin Silva
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France
| | - Sophie Georgin-Lavialle
- AP-HP, Tenon Hospital, Department of Internal Medicine, Paris, France; Sorbonne Université, Centre de Recherche Saint-Antoine (CRSA) INSERM UMRS-938, USA; National French Reference Centre for Auto-inflammatory Diseases and Inflammatory Amyloidosis (CEREMAIA), France
| | - Samuel Deshayes
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| | - Achille Aouba
- Department of Clinical Immunology and Internal Medicine, CHU of Caen Normandie, 14000 Caen, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, 14000 Caen, France
| |
Collapse
|
27
|
Wang Y, Zhao R, Liang Q, Ni S, Yang M, Qiu L, Ji J, Gu Z, Dong C. Organ-based characterization of B cells in patients with systemic lupus erythematosus. Front Immunol 2025; 16:1509033. [PMID: 39917309 PMCID: PMC11798990 DOI: 10.3389/fimmu.2025.1509033] [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: 10/10/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic, inflammatory, and progressive autoimmune disease. The unclear pathogenesis, high heterogeneity, and prolonged course of the disease present significant challenges for effective clinical management of lupus patients. Dysregulation of the immune system and disruption of immune tolerance, particularly through the abnormal activation of B lymphocytes and the production of excessive autoantibodies, lead to widespread inflammation and tissue damage, resulting in multi-organ impairment. Currently, there is no systematic review that examines the specificity of B cell characteristics and pathogenic mechanisms across various organs. This paper reviews current research on B cells in lupus patients and summarizes the distinct characteristics of B cells in different organs. By integrating clinical manifestations of organ damage in patients with a focus on the organ-specific features of B cells, we provide a new perspective on enhancing the efficacy of lupus-targeted B cell therapy strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
28
|
Luo Z, Zhang T, Wang P, Yuan D, Jin S, Di J, Ma R, Yang L, Wang X, Liu J. Activation of V-Domain Immunoglobulin Suppressor of T-Cell Activation by Baloxavir Marboxil Ameliorates Systemic Lupus Erythematosus through Inhibiting Lysophosphatidylcholine/CD40 Ligand. Chem Res Toxicol 2025; 38:193-205. [PMID: 39772456 DOI: 10.1021/acs.chemrestox.4c00449] [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: 01/11/2025]
Abstract
Deficiency of the V-domain immunoglobulin suppressor of T-cell activation (VISTA) accelerates disease progression in lupus-prone mice, and activation of VISTA shows therapeutic effects in mouse models of a lupus-like disease. Metabolic reprogramming of T cells in systemic lupus erythematosus (SLE) patients is important in regulating T-cell function and disease progression. However, the mechanism by which VISTA affects the immunometabolism in SLE remains unclear. Here, we demonstrated that the deficiency of VISTA promoted the synthesis of the metabolite lysophosphatidylcholine (LPC) using untargeted metabolomics and increased the protein expression of the CD40 ligand (CD40L). Furthermore, baloxavir marboxil (BXM), a small molecule agonist of VISTA, significantly ameliorated autoantibody production, renal damage, and imbalance of immune cell subpopulations in the models of a lupus-like disease in mice (chronic graft-versus-host disease and MRL/MpJ-Faslpr/J mice) possibly by inhibiting LPC synthesis to downregulate CD40L protein expression and inhibiting aberrant activation of noncanonical nuclear factor-κB pathway. Our results indicated that BXM targeting VISTA ameliorated lupus-like symptoms by altering lipid metabolism and CD40L expression, which offers novel mechanisms and a promising therapy for SLE.
Collapse
Affiliation(s)
- Zhijie Luo
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Tingting Zhang
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Penglu Wang
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Dingyi Yuan
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Shasha Jin
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Jianwen Di
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixue Ma
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Yang
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Xinzhi Wang
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Liu
- State Key Laboratory of Natural Medicines, New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
29
|
Gao S, Yang C, Huang B, Yang L, Lu L, Yang H, Li T, Pan Q. Comparative efficacy and safety of different recommended doses of telitacicept in patients with systemic lupus erythematosus in China: a systematic review and meta-analysis. Front Immunol 2025; 15:1472292. [PMID: 39867893 PMCID: PMC11757125 DOI: 10.3389/fimmu.2024.1472292] [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: 07/29/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Background Telitacicept, a new biological agent, was approved in China for treating systemic lupus erythematosus (SLE) in 2021. Its optimal dosing for treating SLE remains unclear. Therefore, the aim of this meta-analysis is to evaluate the efficacy and safety of various telitacicept doses in SLE treatment. Methods PubMed, EMBASE, Cochrane libraries, Web of science, China National Knowledge Infrastructure (CNKI), VIP, Wanfang, and Sinomed were searched for the controlled trials that studied the efficacy and safety of telitacicept on SLE patients from their initiation to April 30, 2024. The analysis included three randomized controlled trials (RCT) with 606 participants. We used fixed-effects models for meta-analyses and the risk ratios (RRs) and corresponding 95% confidence intervals (CIs) to evaluate the effectiveness and safety. Heterogeneity was assessed and quantified using I2. Results All telitacicept dosages (80 mg, 160 mg, 240 mg) significantly improved SLE Responder Index 4 (SRI4) responses compared to the control group (RR = 2.20, 95%CI:1.50-3.21, p < 0.0001; RR = 2.18, 95%CI: 1.82-2.62, p < 0.00001; RR = 2.44, 95%CI: 1.67-3.56, p < 0.00001, respectively). The 80 mg, 160 mg, and 240 mg groups also showed better improvement on SELENA-SLE Disease Activity Index (SELENA-SLEDAI) scores (RR = 1.63, 95%CI: 1.23-2.17, p = 0.0008; RR = 1.72, 95%CI: 1.45-2.04, p < 0.00001; RR = 1.73, 95%CI: 1.30-2.30, p = 0.0002, respectively) and Physician Global Assessment (PGA) scores (RR = 1.25, 95%CI: 1.09-1.44, p = 0.002; RR = 1.39, 95%CI: 1.25-1.55, p < 0.00001; RR = 1.24, 95%CI: 1.09-1.42, p = 0.002, respectively). Furthermore, 160 mg group exhibited higher British Isles Lupus Assessment Group (BILAG) score than the control group (RR = 1.11, 95%CI: 1.01-1.22, p = 0.03). As for security, 160 mg telitacicept group had higher incidence of adverse events (AEs) than the control group (RR = 1.10, 95%CI: 1.03-1.18, p = 0.007). Conclusion Telitacicept combined with standard therapy presents potential benefits but there are certain safety concerns with certain dosages of telitacicept, warranting further investigation for optimal dosing strategies in SLE management. Systematic review registration INPLASY.COM, identifier INPLASY202440101.
Collapse
Affiliation(s)
- Shenglan Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chunlong Yang
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bitang Huang
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non−Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lu Lu
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huiting Yang
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Qingjun Pan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Mohamed Thaha UAB, Wan Mohamad WM, Nik Husain NR, Yusop N, Mohamud R, Wan Ghazali WS. Potential and limitations of IL-37, a cytokine targeted for therapy of systemic lupus erythematosus: A Systematic Review. Int Immunopharmacol 2025; 144:113597. [PMID: 39566387 DOI: 10.1016/j.intimp.2024.113597] [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: 08/25/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by dysregulated immune responses and inflammation. Interleukin-37 (IL-37) is a recently discovered immunomodulatory cytokine with potential anti-inflammatory properties. This systematic review explores the relationship between IL and 37 and SLE disease activity, and evaluates its potential as a therapeutic agent. METHODS Electronic databases were searched for studies investigating IL-37 and SLE. Data on IL-37 levels, SLE Disease Activity Index (SLEDAI) score, genetic polymorphisms, and its therapeutic effects from pre-clinical studies were extracted. RESULTS Previous studies presented conflicting findings on IL-37 levels in SLE patients. Some reported positive correlations with disease activity, while others observed associations between lower IL-37 and increased activity. Genetic variations in the IL-37 gene linked to SLE susceptibility have been reported. Pre-clinical studies using engineered mesenchymal stem cells or direct IL-37 treatment showed promise in reducing disease severity in mouse models and cell cultures of SLE. The analysis of multiple studies reveals that IL-37 expression varies significantly across different SLE subtypes. CONCLUSIONS While a potential link exists between IL and 37 and disease activity, genetic predisposition, and therapeutic benefit, further research is needed. Future studies with standardized designs, larger and more diverse populations, and mechanistic investigations are crucial to determine the therapeutic potential of IL-37 for SLE. This review highlights the need for well-designed clinical trials to evaluate the safety and efficacy of IL-37 therapy in patients with SLE.
Collapse
Affiliation(s)
- Ummul Aqeela Balqees Mohamed Thaha
- School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia; Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Wan Majdiah Wan Mohamad
- School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia; Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia.
| | | | - Norhayati Yusop
- School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia; Hospital Universiti Sains Malaysia, USM Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | | |
Collapse
|
31
|
Zheng D, Li X, Wang P, Zhu Q, Huang Z, Zhao T. Exploring the shared mechanism of fatigue between systemic lupus erythematosus and myalgic encephalomyelitis/chronic fatigue syndrome: monocytic dysregulation and drug repurposing. Front Immunol 2025; 15:1440922. [PMID: 39845969 PMCID: PMC11752880 DOI: 10.3389/fimmu.2024.1440922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025] Open
Abstract
Background SLE and ME/CFS both present significant fatigue and share immune dysregulation. The mechanisms underlying fatigue in these disorders remain unclear, and there are no standardized treatments. This study aims to explore shared mechanisms and predict potential therapeutic drugs for fatigue in SLE and ME/CFS. Methods Genes associated with SLE and ME/CFS were collected from disease target and clinical sample databases to identify overlapping genes. Bioinformatics analyses, including GO, KEGG, PPI network construction, and key target identification, were performed. ROC curve and correlation analysis of key targets, along with single-cell clustering, were conducted to validate their expression in different cell types. Additionally, an inflammation model was established using THP-1 cells to simulate monocyte activation in both diseases in vitro, and RT-qPCR was used to validate the expression of the key targets. A TF-mRNA-miRNA co-regulatory network was constructed, followed by drug prediction and molecular docking. Results Fifty-eight overlapping genes were identified, mainly involved in innate immunity and inflammation. Five key targets were identified (IL1β, CCL2, TLR2, STAT1, IFIH1). Single-cell sequencing revealed that monocytes are enriched with these targets. RT-qPCR confirmed significant upregulation of these targets in the model group. A co-regulatory network was constructed, and ten potential drugs, including suloctidil, N-Acetyl-L-cysteine, simvastatin, ACMC-20mvek, and camptothecin, were predicted. Simvastatin and camptothecin showed high affinity for the key targets. Conclusion SLE and ME/CFS share immune and inflammatory pathways. The identified key targets are predominantly enriched in monocytes at the single-cell level, suggesting that classical monocytes may be crucial in linking inflammation and fatigue. RT-qPCR confirmed upregulation in activated monocytes. The TF-mRNA-miRNA network provides a foundation for future research, and drug prediction suggests N-Acetyl-L-cysteine and camptothecin as potential therapies.
Collapse
Affiliation(s)
- Daisi Zheng
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaolong Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peicheng Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingmiao Zhu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiyan Huang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
32
|
Liu Q, Liu Y, Feng H, Zhao L, Wan T. Exploring genetic associations in systemic lupus erythematosus through Mendelian randomization: implications for novel biomarkers and therapeutic targets. Clin Rheumatol 2025; 44:193-205. [PMID: 39126578 DOI: 10.1007/s10067-024-07094-0] [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: 05/22/2024] [Revised: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by a significant health burden. There is an essential need for novel biomarkers and therapeutic targets to improve diagnosis and management. Mendelian randomization (MR) was applied to explore causal links between SLE and various biomarkers like immune cells, metabolites, and inflammatory cytokines using multiple databases. Initially, biomarkers significantly associated with SLE were identified. Bidirectional MR helped clarify these relationships, and a two-step mediation MR examined their effects on SLE risk. Intersection analysis was used to identify biomarkers with consistent effects across datasets. Four biomarkers were identified as having significant associations with SLE risk: 1-palmitoyl-2-arachidonoyl-GPI levels [odds ratio (OR), 1.379; 95% confidence interval (CI), 1.180 to 1.613; FDR, 0.046], IL-17A levels (OR, 2.197; 95% CI, 1.412 to 3.418; FDR, 0.044), N-acetyl-aspartyl-glutamate (NAAG) levels (OR, 0.882; 95% CI, 0.831 to 0.936; FDR, 0.030), and ribitol levels (OR, 0.743; 95% CI, 0.644 to 0.857; FDR, 0.012). Bidirectional MR showed an inverse effect of NAAG on IL-17A levels (OR, 0.978; 95% CI, 0.962 to 0.994; p = 0.006). Mediation analysis indicated that NAAG influenced SLE risk both directly (beta = - 0.108) and indirectly through IL-17A (beta = - 0.018), highlighting the potential mediating role of IL-17A. After expanding the significance criteria to p < 0.05, intersection analysis across multiple datasets revealed 29 biomarkers with consistent beta directions, including 19 potential risk factors (beta > 0) and 10 protective factors (beta < 0) for SLE. This research has revealed significant genetic associations with SLE and demonstrated that IL-17A mediates the relationship between NAAG levels and SLE risk, highlighting potential new targets for personalized therapeutic interventions. Key Points • This study employs MR to identify significant genetic associations between various biomarkers and SLE, providing novel insights into potential biomarkers and therapeutic targets. • Four key biomarkers were identified as significantly associated with SLE risk: 1-palmitoyl-2-arachidonoyl-GPI, IL-17A, N-acetyl-aspartyl-glutamate (NAAG), and ribitol. • The findings suggest that NAAG levels have a protective effect against SLE, partly mediated through IL-17A, indicating a complex interplay between these biomarkers in the pathogenesis of SLE. • Intersectional analysis across multiple datasets revealed 29 biomarkers with consistent effects on SLE risk, highlighting new directions for future research and potential personalized therapeutic strategies.
Collapse
Affiliation(s)
- Qi Liu
- Department of Hepatobiliary Surgery, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
- Medical School of Chinese People's Liberation Army, Beijing, China
| | - Yuyang Liu
- Department of Neurosurgery, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Hui Feng
- Department of Zhantansi Outpatient, Jingzhong Medical District of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lin Zhao
- Department of Endocrinology, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Tao Wan
- Department of Hepatobiliary Surgery, 8th Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
33
|
Wangriatisak K, de Vries C, Sharma RK, Huang W, Grönwall C, Pisitkun P, Gunnarsson I, Malmström V, Chootong P, Faustini F. Association between peripheral activated naive and double negative 2 B-cell subsets and clinical parameters in lupus nephritis patients. Scand J Immunol 2025; 101:e13427. [PMID: 39592449 PMCID: PMC11631828 DOI: 10.1111/sji.13427] [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: 07/31/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024]
Abstract
Altered composition of B-cell compartments is a known feature in patients with systemic lupus erythematosus (SLE). However, deep characterisation of B-cell subsets and their relation to clinical manifestations and disease activity in patients is limited. In this study, we analysed peripheral B-cell subsets phenotype in SLE (n = 35) and healthy controls (HCs, n = 15) by spectral flow cytometry. Disease activity was stratified as inactive (SLEDAI-2 K score 0, n = 2), mild (SLEDAI-2 K score 1-5, n = 12), moderate (SLEDAI-2 K score 6-10, n = 6) or high (SLEDAI-2 K > 10, n = 15). An elevated proportion of activated naive (aNAV), double negative 2 (DN2) and plasmablasts (PB) was observed in patients with high disease activity, compared to other groups of patients and HCs. An upregulation of BTLA was found on both aNAV and DN2 and shifted to lower levels with increasing disease activity. In lupus nephritis (LN) patients (n = 21), aNAV B-cells were especially expanded and positively correlated with DN2 (r = 0.5, p = 0.019) and PB (r = 0.43, p = 0.048). Also, correlation was observed between DN2 and PB (r = 0.6, p = 0.003). Moreover, aNAV frequencies positively correlated with SLEDAI-2 K score, and negatively with the complement fractions C3 and C4. Further, aNAV, DN2 and PB were more expanded in association with positive anti-dsDNA antibodies, rather than other antibody specificities (anti-Sm). These data suggest roles of extrafollicular B cells as key players in disease development of LN. Their association with presence of anti-dsDNA antibodies may indicate their value as candidate biomarkers of kidney involvement in SLE.
Collapse
Affiliation(s)
- Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol UniversityNakhon PathomThailand
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Charlotte de Vries
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Wenqi Huang
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Caroline Grönwall
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Iva Gunnarsson
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Medicine Unit Dermatology, Gastroenterology, Rheumatology; Unit of RheumatologyKarolinska University Hospital SolnaStockholmSweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Center for Molecular MedicineKarolinska InstitutetStockholmSweden
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical TechnologyMahidol UniversityNakhon PathomThailand
| | - Francesca Faustini
- Division of Rheumatology, Department of MedicineKarolinska Institutet, Karolinska University Hospital SolnaStockholmSweden
- Medicine Unit Dermatology, Gastroenterology, Rheumatology; Unit of RheumatologyKarolinska University Hospital SolnaStockholmSweden
| |
Collapse
|
34
|
Aljumaili KMH, Haghi M, Khalaj Kondori M, Ardalan MR, Hosseinpour Feizi MA. Association Study of rs1632947, rs1233334, and rs371194629 Polymorphisms in Human Leukocyte Antigen G Gene Expression and soluble Human Leukocyte Antigen G with Lupus. DNA Cell Biol 2025; 44:25-31. [PMID: 39503744 DOI: 10.1089/dna.2024.0144] [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: 01/12/2025] Open
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease that has been associated with human leukocyte antigen G (HLA-G) in previous studies on immunological diseases. This study aimed to investigate the association between three HLA-G gene polymorphisms (rs1632947, rs1233334, and rs371194629) and their impact on HLA-G mRNA expression and soluble HLA-G levels in serum. Genotyping was performed using TaqMan probe PCR. RNA extraction, reverse transcription PCR, and real-time PCR assays were conducted to assess the expression of the HLA-G gene in tissue samples. Soluble HLA-G was measured using enzyme-linked immunosorbent assay in serum. Results show a significant difference in the frequency of the G allele for two 5'-untranslated region (UTR) polymorphisms of the HLA-G gene (rs1632947 and rs1233334) located at positions -964 and -725, respectively, between lupus patients and controls, with p-values of 0.009 and 0.040, respectively. In addition, the study identified the 14 bp insertion allele of the rs371194629 polymorphism located in the 3' UTR of the gene as a risk factor for lupus, with a p-value of 0.001. Our results also indicate that lupus-related alleles may increase the risk of developing the disease by upregulating the expression of HLA-G and increasing soluble HLA-G levels in serum. The findings of the study suggest that the identified genetic variants may play a role in the development of lupus and could be useful in identifying individuals at risk for the disease. These results are important for advancing our understanding of the genetic basis of lupus and may have implications for the development of new treatments and diagnostic tools for the disease.
Collapse
Affiliation(s)
| | - Mehdi Haghi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | | |
Collapse
|
35
|
Du J, Luo H, Ye S, Zhang H, Zheng Z, Liu K. Unraveling IFI44L's biofunction in human disease. Front Oncol 2024; 14:1436576. [PMID: 39737399 PMCID: PMC11682996 DOI: 10.3389/fonc.2024.1436576] [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: 05/22/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Interferon-induced protein 44-like (IFI44L) is regarded as an immune-related gene and is a member of interferon-stimulated genes (ISGs). They participate in network transduction, and its own epigenetic modifications, apoptosis, cell-matrix formation, and many other pathways in tumors, autoimmune diseases, and viral infections. The current review provides a comprehensive overview of the onset and biological mechanisms of IFI44L and its potential clinical applications in malignant tumors and non-neoplastic diseases.
Collapse
|
36
|
Ding Q, Zhou Y, Feng Y, Sun L, Zhang T. Bruton's tyrosine kinase: A promising target for treating systemic lupus erythematosus. Int Immunopharmacol 2024; 142:113040. [PMID: 39216117 DOI: 10.1016/j.intimp.2024.113040] [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: 07/08/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disorder involving multiple organs and systems. There is growing evidence that autoreactive B cells occupy a central role in the occurrence and progression of SLE due to their ability to generate pathogenic autoantibodies. Small molecule inhibitors targeting Bruton's tyrosine kinase (BTK), a crucial intracellular kinase regulating B cell development and function, emerge as a new strategy to treat SLE in recent years and are superior to biologic agents depleting B cells in many aspects. Supportive data obtained from lupus-prone mice preliminarily demonstrated the promising therapeutic potential of BTK inhibition. However, these BTK inhibitors, including elsubrutinib, evobrutinib, etc., mostly face with unsatisfactory efficacy and certain safety issues during clinical use, driving the quest for new-generation inhibitors with improved potency and higher selectivity. This paper elaborates the importance of BTK involvement in SLE pathogenesis, reviews the clinical research progress of BTK inhibitors for SLE and discusses limitations and challenges the drugs met in development, in order to contribute to a deeper understanding of disease mechanism and provide a reference for new-generation BTK inhibitor research.
Collapse
Affiliation(s)
- Qiaoyi Ding
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yifan Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lan Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
37
|
Jiang W, Xu Y, Yin Q. Graft survival and mortality outcomes after kidney transplant in patients with lupus nephritis: a systematic review and meta-analysis. Ren Fail 2024; 46:2296000. [PMID: 38178546 PMCID: PMC10773647 DOI: 10.1080/0886022x.2023.2296000] [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: 09/11/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
To explore the effect of lupus nephritis (LN) on graft survival in renal transplant patients. Literature search was conducted in PubMed, EMBASE and Scopus database for randomized controlled trials (RCTs), cohort, and case-control studies. The target population of interest was adult patients (aged >18 years) with end-stage renal disease (ESRD) and no history of previous renal transplants. Primary outcomes of interest were graft survival and patient survival. Pooled effect estimates were calculated using random-effects models and reported as hazard ratio (HR) with 95% confidence intervals (CI). A total of 15 studies were included. Compared to patients with ESRD due to other causes, patients with LN undergoing kidney transplant had lower patient survival rate (HR 1.15, 95% CI: 1.01, 1.31; N = 15, I2=34.3%) and worse graft survival (HR 1.06, 95% CI: 1.01, 1.11; N = 16, I2=0.0%), especially when studies with deceased donor were pooled together. Studies with a larger sample size (>200) showed that LN was strongly associated with lower graft and patient survival rates. Elevated risk of mortality in LN patients was detected in case-control studies, but not RCTs. On the other hand, RCTs, but not case-control studies, showed an increased risk of poor graft survival in LN patients. The findings suggest that the presence of LN might have a negative impact on both the graft survival and the overall patient survival of post-transplant ESRD patients. Further studies that account for factors such as study methodology, donor characteristics, and sample size are needed to reach definitive conclusions. Renal transplant patients with LN should undergo regular follow-up examinations.
Collapse
Affiliation(s)
- Weizhong Jiang
- Department of Nephrology, Huzhou Central Hospital, Affiliated Central Hospital HuZhou University, Huzhou City, Zhejiang Province, China
| | - Yunfen Xu
- Department of Nephrology, Huzhou Central Hospital, Affiliated Central Hospital HuZhou University, Huzhou City, Zhejiang Province, China
| | - Qichun Yin
- Department of Nephrology, Huzhou Central Hospital, Affiliated Central Hospital HuZhou University, Huzhou City, Zhejiang Province, China
| |
Collapse
|
38
|
Mora VP, Quero FB, Troncoso-Bravo T, Orellana C, Pereira P, Mackern-Oberti JP, Funes SC, Soto JA, Bohmwald K, Bueno SM, Kalergis AM. Partial long-term clinical improvement after a BCG challenge in systemic lupus erythematosus-prone mice. Autoimmunity 2024; 57:2380465. [PMID: 39034498 DOI: 10.1080/08916934.2024.2380465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/07/2024] [Indexed: 07/23/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune disorder that causes a breakdown of immune tolerance. Current treatments mainly involve general immunosuppression, increasing the risk of infections. On the other hand, Bacillus Calmette-Guérin (BCG) has been investigated as a potential therapy for autoimmune diseases in recent years, prompting an ongoing investigation. This study aimed to evaluate the effect of BCG vaccination on early and late clinical presentation of SLE in a murine disease model. MRL/MPJ-Faslpr mice were immunized with BCG or treated with PBS as a control. The progress of the disease was evaluated at 27 days post-immunization (dpi) (early) and 56 dpi (late). Clinical parameters and proteinuria were monitored. Blood samples were collected for measurement of antinuclear antibodies (ANAs), anti-double-stranded DNA (anti-dsDNA), and cytokine determination was performed using ELISA. Samples collected from mice were analyzed by flow cytometry and histopathology. We observed a clinical improvement in BCG-treated mice, reduced proteinuria in the latter stages of the disease, and decreased TNF-α. However, BCG did not elicit significant changes in ANAs, anti-dsDNA, histopathological scores, or immune cell infiltration. BCG was only partially beneficial in an SLE mouse model, and further research is needed to determine whether the immunity induced by this vaccine can counteract lupus's autoimmune response.
Collapse
Affiliation(s)
- Valentina P Mora
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Francisco B Quero
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tays Troncoso-Bravo
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ingeniería Química y Bioprocesos, Facultad de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Orellana
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Pereira
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan P Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo, CONICET, Mendoza, Argentina
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Samanta C Funes
- Instituto Multidisciplinario de Investigaciones Biológicas-San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de San Luis (UNSL), San Luis, Argentina
| | - Jorge A Soto
- Millennium Institute of Immunology and Immunotherapy. Departamento de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Karen Bohmwald
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
39
|
Nieves C, Victoria da Costa Ghignatti P, Aji N, Bertagnolli M. Immune Cells and Infectious Diseases in Preeclampsia Susceptibility. Can J Cardiol 2024; 40:2340-2355. [PMID: 39304126 DOI: 10.1016/j.cjca.2024.09.012] [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: 06/02/2024] [Revised: 08/26/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
Preeclampsia is a severe pregnancy disorder, affecting approximately 10% of pregnancies worldwide, characterised by hypertension and proteinuria after the 20th week of gestation. The condition poses significant risks to both maternal and fetal health, including cardiovascular complications and impaired fetal development. Recent trends indicate a rising incidence of preeclampsia, correlating with factors such as advanced maternal age and cardiovascular comorbidities. Emerging evidence also highlights a notable increase in the association between autoimmune and infectious diseases with preeclampsia. Autoimmune conditions, such as type 1 diabetes and systemic lupus erythematosus, and infections triggered by global health challenges, including leptospirosis, Zika, toxoplasmosis, and Chagas disease, are now recognised as significant contributors to preeclampsia susceptibility by affecting placental formation and function. This review focuses on the immunologic mechanisms underpinning preeclampsia, exploring how immune system dysregulation and infectious triggers exacerbate the condition. It also discusses the pathologic mechanisms, including galectins, that preeclampsia shares with autoimmune and infectious diseases, and their significant risk for adverse pregnancy outcomes. We emphasise the necessity for accurate diagnosis and vigilant monitoring of immune and infectious diseases during pregnancy to optimise management and reduce risks. By raising awareness about these evolving risks and their impact on pregnancy, we aim to enhance diagnostic practices and preventive strategies, ultimately improving outcomes for pregnant women, especially in regions affected by environmental changes and endemic diseases.
Collapse
Affiliation(s)
- Cecilia Nieves
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada.
| | - Paola Victoria da Costa Ghignatti
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Narjiss Aji
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Mariane Bertagnolli
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
40
|
Barlianto W, Wulandari D, Sari TL, Rachmaningrum RR, Asasain RI. Effects of Nigella sativa on disease activity, T lymphocytes and inflammatory cytokine profiles in pediatric systemic lupus erythematosus: A randomized controlled trial. NARRA J 2024; 4:e1063. [PMID: 39816053 PMCID: PMC11731933 DOI: 10.52225/narra.v4i3.1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/01/2024] [Indexed: 01/30/2025]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with diverse manifestations, requiring long-term treatment that can have side effects, particularly in pediatric patients. Nigella sativa has shown potential for improving SLE symptoms due to its anti-inflammatory and immunomodulatory effects. The aim of this study was to investigate the immunomodulatory effect of N. sativa oil (NSO) on disease activity, T lymphocyte activity and inflammatory cytokine profiles in pediatric SLE patients. A randomized, double-blinded, placebo-controlled clinical trial was conducted at Saiful Anwar Hospital in Malang, Indonesia. Pediatric patients with SLE were randomly assigned to receive either one gram of NSO or a placebo containing starch in capsule form as adjunct therapy alongside their SLE primary treatment. Blood samples were collected before treatment and after eight weeks of daily capsules. Disease activity was assessed using the SLE Disease Activity Index 2000 (SLEDAI-2 K); flow cytometry was used to identify T helper lymphocytes, and serum cytokine levels were measured using ELISA. The statistical analysis tests were performed to compare the outcomes between groups at baseline or after the treatment, and within-group comparisons before and after the study period, as appropriate. A total of 32 patients were included in the study. A significant decrease in the SLEDAI-2 K score was observed at post-treatment in both the NSO and placebo groups (p < 0.001 and p = 0.025, respectively). The percentage of T helper 17 (Th17) cells was significantly reduced in both the NSO and placebo groups post-treatment compared to pre-treatment (p = 0.026 and p = 0.034, respectively). Conversely, the post- treatment percentage of regulatory T (Treg) cells increased significantly in both groups. A significant reduction in interleukin (IL)-2 levels was observed in the NSO and placebo groups at post-treatment compared to pre-treatment (p = 0.006 and p = 0.046, respectively). Additionally, there were increases in IL-4 and IL-6 serum levels in both groups at post-treatment compared to pre-treatment (p < 0.05). This study highlights that although disease activity was not significantly different between NSO and placebo groups, NSO could affect the inflammatory cytokine profiles in pediatric SLE patients.
Collapse
Affiliation(s)
- Wisnu Barlianto
- Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Desy Wulandari
- Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Tita L. Sari
- Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | | | - Rayi I. Asasain
- Department of Pediatrics, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
41
|
Ginting AR, Munir D, Amin MM, Darlan DM, Putra A, Rusda M, Mutiara E, Mayasari E, Rozi MF. Mesenchymal stem cells for immune modulation in systemic lupus erythematosus: From bench research to clinical applications. NARRA J 2024; 4:e994. [PMID: 39816093 PMCID: PMC11731813 DOI: 10.52225/narra.v4i3.994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/15/2024] [Indexed: 01/18/2025]
Abstract
Systemic lupus erythematosus (SLE) is a prevalent autoimmune disease affecting multiple organ systems. Disease progression is inevitable as part of its natural course, necessitating aggressive therapeutic strategies, particularly with the use of immunosuppressants. Long-term use of steroids and other immunosuppressants is associated with significant adverse effects. Mesenchymal stem cells (MSCs) have been shown to modulate the immune response, leading to immunosuppressive effects against self-antigens. MSCs have demonstrated the ability to modulate several immune cell populations, contributing to favorable outcomes in controlling immune and inflammatory conditions. Recent evidence has shown an increase in Treg and Breg cell subsets following MSC administration, along with modulation of other immune cells, including dendritic cells, B cells, and T cells. However, the balance between MSC pro-inflammatory and anti-inflammatory phenotypic activation remains a critical factor in determining therapeutic outcomes. Various covariates also influence the efficacy of MSC therapy. The aim of this study was to provide a comprehensive overview of the utilization of mesenchymal stem cells (MSCs) in SLE treatment, leveraging their immunomodulatory and immunosuppressive capabilities. Understanding the fundamental preclinical effects of MSCs and recent findings from clinical studies may enhance the potential of MSC therapy in the management of SLE patients.
Collapse
Affiliation(s)
- Andi R. Ginting
- Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Delfitri Munir
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Pusat Unggulan Iptek Tissue Engineering, Universitas Sumatera Utara, Medan, Indonesia
| | - Mustafa M. Amin
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Psychiatry, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Dewi M. Darlan
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Parasitology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research Center, Semarang, Indonesia
- Faculty of Postgraduate Biomedical Science, Universitas Islam Sultan Agung, Semarang, Indonesia
| | - Muhammad Rusda
- Department of Doctoral Program in Medical Sciences, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Erna Mutiara
- Faculty of Public Health, Universitas Sumatera Utara, Medan, Indonesia
| | - Evita Mayasari
- Department of Microbiology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad F. Rozi
- Department of Internal Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
42
|
Zhao Y, Li X, Wang L, Xu W, Liu B, Yan S, Ma Z, Yang Q. Phenotypes of patients with lupus-associated pulmonary hypertension in the Chinese Han population: a cluster analysis. Clin Rheumatol 2024; 43:3713-3722. [PMID: 39485555 DOI: 10.1007/s10067-024-07216-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/30/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE Pulmonary hypertension (PH) is a severe complication of systemic lupus erythematosus (SLE). This study investigated the clinical features of patients with SLE-PH in China based on disease manifestations and organ involvement to define precise treatment of SLE and early prevention of complications. METHODS In total, 205 SLE-PH patients were included in this study. A cluster analysis was applied according to six clinical and serological variables to define different subgroups of patients. The survival rates of SLE-PH patients and risk factors that influenced prognosis were also compared and a clinical prediction model was developed for the diagnosis of associated lupus nephritis (LN). RESULTS Patients were clustered in five groups. Patients in cluster 1 had severe renal damage (all patients had LN and had the highest creatinine and urea nitrogen and the lowest eGFR levels). Patients in cluster 2 had mild renal damage (more than half of the patients had associated LN and 87.5% had increased urinary protein levels but presented a lower degree of renal damage than those in the first group. Patients in cluster 3 had low-grade proteinuria (all patients had 24h urinary protein < 0.5g). Patients in cluster 4 had hematuria or urinary tubular damage (26% of patients had associated LN, but none of the patients had proteinuria. In cluster 5, patients barely had any major organ involvement. The clinical prediction model for a diagnosis of SLE-PH-LN was anti-dsDNA antibodies > 364.64 IU/mL and neutrophil-to-leukocyte ratio (NLR) > 5.55. CONCLUSION Our findings provide evidence indicating that SLE-PH presents varying clinical phenotypes and the treatment varies accordingly, suggesting the need for individualized treatment. Key Points • Clustered grouping of patients with SLE-PH is associated with renal injury. • This may be because PH and LN share a common pathogenesis. • The clinical prediction model for a diagnosis of SLE-PH-LN was anti-dsDNA antibodies >364.64 IU/mL and NLR >5.55.
Collapse
Affiliation(s)
- Yaqi Zhao
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xinya Li
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Liya Wang
- Heze Medical College, Heze, Shandong, China
| | - Wei Xu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Baocheng Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Suyan Yan
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
43
|
Luan Y, Min Q, Zhang R, Wen Z, Meng X, Hu Z, Feng X, Yu M, Dong L, Wang JY. EAF2 deficiency attenuates autoimmune disease in Fas lpr mice by modulating B cell activation and apoptosis. iScience 2024; 27:111220. [PMID: 39555413 PMCID: PMC11565555 DOI: 10.1016/j.isci.2024.111220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/06/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
MRL/lpr mice develop systemic lupus erythematosus-like autoimmunity due to defective FAS-mediated apoptosis. We generated Fas lpr mice deficient in EAF2, a transcription elongation-associated factor known to promote apoptosis in germinal center (GC) B cells and crucial for preventing autoimmunity. Contrary to expectations, EAF2 deficiency significantly reduced lymphadenopathy and splenomegaly, extended lifespan, and alleviated nephritis by decreasing renal immune complex deposition. Additionally, EAF2 deficiency markedly reduced accumulation of activated B cells, GC B cells, plasma cells, and the abnormal B220+CD3+ T cells in Fas lpr mice. Further analysis revealed that Eaf2 -/- Fas lpr B cells showed hyperactivation upon various stimulations, followed by increased death. RNA sequencing of the B220+CD3+ cells revealed a downregulation in survival-promoting genes such as Bcl-2 and Akt and an upregulation of proapoptotic genes. We conclude that the combined deficiency in FAS- and EAF2-mediated apoptotic pathways leads to B cell hyperactivation and subsequent death, thereby ameliorating systemic autoimmunity in this model.
Collapse
Affiliation(s)
- Yingying Luan
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Min
- Department of Clinical Immunology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| | - Runyun Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zichao Wen
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Feng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Clinical Immunology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
| | - Lulu Dong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Clinical Immunology, Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai, China
| |
Collapse
|
44
|
Ren Y, Tian J, Shi W, Feng J, Liu Y, Kang H, He Y. Evaluation of ocular surface inflammation and systemic conditions in patients with systemic lupus erythematosus: a cross-sectional study. BMC Ophthalmol 2024; 24:492. [PMID: 39533209 PMCID: PMC11556210 DOI: 10.1186/s12886-024-03760-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE The cross-sectional study was designed to evaluate the association of ocular surface inflammation with systemic conditions in patients with systemic lupus erythematosus (SLE). METHODS The study enrolled 30 SLE patients and 30 controls. Ocular symptoms were evaluated using the Ocular Surface Disease Index (OSDI) questionnaire. Tear samples from all participants were collected for tear multi-cytokine and chemokine concentration analysis. All participants were assessed for dry eye disease (DED), including Schirmer I test, tear break-up time (TBUT), corneal fluorescein staining (CFS), meibomian gland secretion (MGS), lid-parallel conjunctival folds (LIPCOF), corneal clarity, and symblepharon. Besides, all participants were also examined for conjunctival impression cytology to measure the density of conjunctival goblet cells (CGCs). The peripheral blood indicators from SLE patients were also collected to measure the SLE-associated autoantibody specificities and systemic inflammatory indicators. Pearson and Spearman's analysis were uesd to examine the correlation between tear cytokines, CGCs, DED-related indicators, and systemic conditions. RESULTS The two groups were matched for age and gender in this study. 36.67% of eyes (11 in 30) of SLE patients and 13.33% of eyes (4 in 30) of controls were diagnosed with DED. OSDI scores, abnormal TBUT percentages, CFS percentages, and DED grading were all higher in SLE patients than in control group, while density of CGCs was lower. There were no significant differences in Schirmer I test, MGS, LIPCOF, corneal clarity, and symblepharon between SLE patients and controls. The levels of tear chemokine (C-X-C motif) ligand 11 (CXCL11) and cytokine interleukin-7 (IL-7) in patients with SLE were significantly higher than those in control group. Moreover, among SLE patients, the severity of DED and the level of tear chemokine CXCL11 were significantly positively correlated with SLE-associated autoantibody specificities. CONCLUSION Dry eye and tear cytokines and chemokines-mediated ocular surface inflammation persist in SLE patients and are associated with systemic conditions. Therefore, it is necessary for patients with SLE to combine systemic and ocular assessments.
Collapse
Affiliation(s)
- Yuerong Ren
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jing Tian
- Department of Rheumatology and Immunology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wen Shi
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Jianing Feng
- Xi'an People's Hospital (Xi'an Fourth Hospital), Shaanxi Eye Hospital, Northwest University Affiliated People's Hospital, Xi'an, Shaanxi Province, China
| | - Yingyi Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Huanmin Kang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, China
| | - Yan He
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmic and Visual Science Key Laboratory, Beijing, China.
| |
Collapse
|
45
|
Liu Y, Yang X. A review on the novel biomarkers of systemic lupus erythematosus discovered via metabolomic profiling. Front Immunol 2024; 15:1443440. [PMID: 39569194 PMCID: PMC11576423 DOI: 10.3389/fimmu.2024.1443440] [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: 06/24/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disease affecting various body organs and systems. The diagnosis of SLE and its complications is based on evident clinical symptoms, serological marker levels, and pathological findings. Some serological markers have a low sensitivity and specificity, and biopsy procedures are invasive in nature. Hence, metabolomics has emerged as a valuable tool for SLE screening and categorization. Its application has contributed significantly to identifying SLE pathogenesis, improving clinical diagnosis, and developing treatment approaches. This review provides an overview of the utilization of metabolomics in the study of SLE, focusing on advancements in understanding the disease's pathogenesis, aiding in diagnosis, and monitoring treatment efficacy.
Collapse
Affiliation(s)
- Yinghong Liu
- Department of Rheumatology, Chongqing University Central Hospital, Chongqing, China
- Department of Rheumatology, Chongqing Emergency Medical Center, Chongqing, China
| | - Xiaojuan Yang
- Department of Rheumatology, Chongqing University Central Hospital, Chongqing, China
- Department of Rheumatology, Chongqing Emergency Medical Center, Chongqing, China
| |
Collapse
|
46
|
Sharma Y, Bala K. Multifarious Aspect of Cytokines as an Immuno-Therapeutic for Various Diseases. J Interferon Cytokine Res 2024; 44:477-485. [PMID: 39394036 DOI: 10.1089/jir.2024.0090] [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: 10/13/2024] Open
Abstract
Cytokines are known to be a group of growing small proteins that are majorly responsible for the transmission of signals and communication between hematopoietic cells, the cells of the human immune system, and other types of cells. Cytokines play a dominant role in different types of disorders and in perpetuating the inflammation-related disorders. The production of cytokines is a natural process inside the body of a human being against any foreign invasion or due to some pathogenic state to maintain the homeostasis. Cytokines respond in two ways; in some cases, the production and development of cytokines as a therapeutic discovery or intervention will enhance the treatment process and support the reaction given by the body against any pathogenic activity, and in some cases, overproduction of these cytokines responds in the opposite way and behaves as antagonists toward a typical therapeutic drug and its treatment. Overall, 41 articles were reviewed, and it was found that cytokines have proved to be a therapeutic approach among various diseases and can be utilized as a good candidate or a better choice for cancer therapeutics in future development.
Collapse
Affiliation(s)
- Yash Sharma
- Department of Biotechnology, IILM University, Greater Noida, India
| | - Kumud Bala
- Department of Biotechnology, IILM University, Greater Noida, India
- Therapeutics and Molecular Diagnostic Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India
| |
Collapse
|
47
|
Varaganti V, Vadakedath S, Ca J, Kandi V, B PV, Hussain MH, V A, Gayathri K. Mechanisms Underlying Gender Influence on the Clinical Course and Immunopathogenesis of Systemic Lupus Erythematosus: An Explorative Review. Cureus 2024; 16:e73646. [PMID: 39677179 PMCID: PMC11645479 DOI: 10.7759/cureus.73646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder with a complex clinical course and diverse presentations. The immunopathogenesis of SLE has long intrigued physicians and researchers. Despite its extensive global prevalence, there is no specific treatment to prevent and treat SLE, and in the majority of SLE patients, the management involves controlling disease remissions and symptom reactivations or flares. SLE patients suffer from damage to different organs of the body, complicating disease management. They are predisposed to infectious diseases that could contribute to enhanced disease progression. Devising effective management strategies requires a comprehensive understanding of the effects of the disease and its influence on the immune system. SLE affects females more frequently than men. However, male SLE patients often suffer from more severe disease than females. Gender variations have also been noted in clinical manifestations in patients with SLE. In light of this, additional research is needed to understand these variations and promote the progress of gender-specific patient management and treatment strategies. This review aimed to compare the influence of gender on the clinical consequences, immunopathogenesis, and associated consequences between male and female SLE patients. An extensive literature search was conducted to collect relevant data. PubMed, MEDLINE, Embase, and Google Scholar were searched from inception to the present for articles that compared clinical outcomes and associated disorders in terms of gender among SLE patients. We also explored the immunopathogenesis, mechanisms underlying gender-based clinical effects of SLE, and infectious disease-related consequences. Additionally, we provide key updates regarding the treatment and management of SLE.
Collapse
Affiliation(s)
- Vamshi Varaganti
- Medicine, Prathima Institute of Medical Sciences, Karimnagar, IND
| | | | - Jayashankar Ca
- Internal Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Venkataramana Kandi
- Clinical Microbiology, Prathima Institute of Medical Sciences, Karimnagar, IND
| | - Pooja V B
- General Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Mir Hyder Hussain
- General Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Anuradha V
- General Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| | - Kalidindi Gayathri
- General Medicine, Vydehi Institute of Medical Sciences and Research Centre, Bangalore, IND
| |
Collapse
|
48
|
Ceobanu G, Edwards CJ. JAK inhibitors in systemic lupus erythematosus: Translating pathogenesis into therapy. Lupus 2024; 33:1403-1415. [PMID: 39383302 DOI: 10.1177/09612033241287594] [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: 10/11/2024]
Abstract
Systemic lupus erythematosus (SLE) is a complex multi-organ autoimmune disease marked by the production of autoantibodies against nuclear structures, formation of immune complexes, and chronic inflammation triggered by their tissular deposition. SLE is characterized by alternating periods of relapse and remission and each flare has the potential to cause new organ damage related to either the disease process or the medication toxicity. Despite remarkable progress across its multiple domains, SLE is still an area with many unmet needs, calling for innovative and practical solutions. The efforts of the drug development programme in lupus have led to considerable growth in the last decade, owing to the approval of belimumab, anifrolumab, and voclosporin. The increasing understanding of the pathogenesis of the disease has enabled the exploration of novel therapeutic strategies. New discoveries in the intricate cytokine kaleidoscope of lupus have made the concept of targeted therapy an attractive and promising research focus. JAK inhibitors are oral targeted therapies approved for a wide variety of diseases across the Rheumatology, Gastroenterology, Dermatology, and Haematology fields. Multiple JAKis are currently being investigated in SLE. This paper aims to summarize existing data coming from both clinical trials and case reports regarding the use of JAK inhibitors in SLE.
Collapse
Affiliation(s)
- Gabriela Ceobanu
- NIHR Southampton Clinical Research Facility, University Hospital Southampton, Southampton, UK
| | - Christopher J Edwards
- NIHR Southampton Clinical Research Facility, University Hospital Southampton, Southampton, UK
| |
Collapse
|
49
|
Nishat SMH, Alzaabi AA, Alzaabi FM, Al Tarawneh DJ, Al Tarawneh YJ, Khan A, Khan MAM, Siddiqui TW, Siddiqui RW, Siddiqui SW. Connecting the Dots: Exploring the Association Between Systemic Lupus Erythematosus and Thyroid Disorders. Cureus 2024; 16:e74469. [PMID: 39726471 PMCID: PMC11669916 DOI: 10.7759/cureus.74469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease marked by chronic inflammation and tissue damage that impacts multiple organ systems and diminishes the quality of life. Among the frequent comorbidities in SLE, thyroid dysfunction, including hypothyroidism and hyperthyroidism, stands out due to its high prevalence and common autoimmune basis. This review examines the epidemiological, genetic, and immunological factors that link SLE with autoimmune thyroid diseases such as Hashimoto's thyroiditis and Graves' disease. These overlapping mechanisms suggest a shared pathophysiological foundation that increases the risk of thyroid dysfunction in SLE patients. Clinically, distinguishing thyroid dysfunction from SLE symptoms, such as fatigue and cognitive difficulties, remains challenging, making regular thyroid screening in SLE patients essential. A multidisciplinary approach, bringing together rheumatologists and endocrinologists, is crucial to provide comprehensive care and improve outcomes for patients managing both conditions.
Collapse
Affiliation(s)
| | - Asma A Alzaabi
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Fatema M Alzaabi
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Dana J Al Tarawneh
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Yusuf J Al Tarawneh
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Abdallah Khan
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | | | - Tabish W Siddiqui
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | - Raqshan W Siddiqui
- Internal Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, ARE
| | | |
Collapse
|
50
|
Priya K, Thacker H, Chaubey M, Rai M, Singh S, Rawat S, Giri K, Mohanty S, Rai G. Dexamethasone and IFN-γ primed mesenchymal stem cells conditioned media immunomodulates aberrant NETosis in SLE via PGE2 and IDO. Front Immunol 2024; 15:1461841. [PMID: 39544931 PMCID: PMC11560778 DOI: 10.3389/fimmu.2024.1461841] [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: 07/09/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Background Systemic Lupus Erythematosus (SLE) is characterized by dysregulated immune responses, with neutrophil extracellular traps (NETs) playing a significant role. NETs are recognized by autoantibodies in SLE patients, exacerbating pathology. Both excessive NET formation and impaired degradation contribute to SLE pathophysiology. Objective To investigate the immunomodulatory effects of Dexamethasone-primed Wharton's jelly (WJ) derived MSCs CM (DW) and IFN-γ-primed WJ-MSCs-CM (IW) on NETosis and associated protein markers in SLE patients' LPS or ribonucleoprotein immune complexes (RNP ICs) induced neutrophils and in pristane induced lupus (PIL) model. And to elucidate the mechanism involved therein. Methods We investigated the immunomodulatory effects of DW and IW on NETosis in SLE. Utilizing ex vivo and in vivo models, we assessed the impact of preconditioned media on NET formation and associated protein markers neutrophil elastase (NE), citrullinated histone (citH3), myeloperoxidase (MPO), cytoplasmic and mitochondrial ROS production. We also examined the involvement of key immunomodulatory factors present in DW and IW, including prostaglandin E2 (PGE2), indoleamine 2,3-dioxygenase (IDO), and transforming growth factor-beta (TGF-β). Results Preconditioned media effectively suppressed NETosis and reduced ROS generation in SLE neutrophils, indicating their immunomodulatory potential. Inhibition studies implicated IDO and PGE2 in mediating this effect. Combined treatment with DW or IW together with hydroxychloroquine (HCQ) demonstrated superior efficacy over HCQ alone, a standard SLE medication. In PIL mouse model, DW and IW treatments reduced NETosis, ROS generation, as evidenced by decreased NET-associated protein expression in vital organs. Conclusion Our study highlights the multifaceted impact of IW and DW on NETosis, ROS dynamics, and lupus severity in SLE. These findings underscore the potential of preconditioned media for the development of targeted, personalized approaches for SLE treatment.
Collapse
Affiliation(s)
- Khushbu Priya
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Hiral Thacker
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Manaswi Chaubey
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Madhukar Rai
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | - Sonali Rawat
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, India
| | - Kiran Giri
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, AIIMS, New Delhi, India
| | - Geeta Rai
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|