101
|
Amer BE, Afifi E, Mouffokes A, Hamad AA, Amin AM, Abdelwahab OA. Does baricitinib reduce disease activity in patients with systemic lupus erythematosus? A systematic review and meta-analysis of randomized controlled trials. Clin Rheumatol 2024; 43:579-589. [PMID: 37581759 DOI: 10.1007/s10067-023-06731-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023]
Abstract
Baricitinib is a selective Janus kinase inhibitor that has recently been approved for treating certain autoimmune disorders. This meta-analysis pooled the conflicting results from all published randomized controlled trials (RCTs) about the efficacy and safety of baricitinib in patients with systemic lupus erythematosus (SLE). We systemically searched four electronic databases. RCTs comparing baricitinib versus placebo were included. Our outcomes were pooled as the risk ratio (RR) in the random effects model. Our primary outcome was the proportion of patients who achieved a SLE Responder Index-4 (SRI-4) response. A total of three RCTs, comprising 1849 patients, were included. Baricitinib 4 mg was associated with a significantly higher proportion of patients who attained SRI-4 response at week 24 (RR = 1.19, 95% CI [1.05, 1.35], P < 0.01). However, this did not reach statistical significance with baricitinib 4 mg at week 52 and baricitinib 2 mg at both week 24 and week 52 (RR = 1.13, 95% CI [0.96, 1.34], P = 0.15; RR = 1.09, 95% CI [0.96, 1.24], P = 0.20; RR = 1.05, 95% CI [0.92, 1.19], P = 0.50, respectively). The risk for serious infections was higher in the baricitinib 4 mg group (RR = 2.23, 95% CI [1.13, 4.37], P = 0.02). Baricitinib 2 mg did not show any clinical benefit. In contrast, baricitinib 4 mg might have the potential to reduce SLE disease activity; however, further research is required to evaluate its long-term efficacy. Until higher-quality evidence is developed, the benefits and risks of baricitinib should be considered before initiating its therapy. Key Points • Baricitinib is a selective Janus kinase inhibitor that has recently been approved for treating certain autoimmune disorders; however, its efficacy in patients with systemic lupus erythematosus (SLE) is still inconclusive. • In our meta-analysis, baricitinib 2 mg did not show any clinical benefit. In contrast, baricitinib 4 mg significantly reduced SLE activity in terms of SRI-4 response at week 24. However, this did not reach statistical significance at week 52. • Further studies are required to investigate the long-term efficacy of baricitinib 4 mg in patients with SLE.
Collapse
Affiliation(s)
- Basma Ehab Amer
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA.
- Faculty of Medicine, Benha University, Benha, Egypt.
| | - Eslam Afifi
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Faculty of Medicine, Benha University, Benha, Egypt
| | - Adel Mouffokes
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Faculty of Medicine, University of Oran, 1 Ahmed Ben Bella, Oran, Algeria
| | - Abdullah Ashraf Hamad
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Ahmed Mostafa Amin
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Omar Ahmed Abdelwahab
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
102
|
McSweeney SM, Mounsey S, Arujuna N, Garibaldinos T, Sarkany R, Fassihi H. A single-centre retrospective review of treatment responses and adverse events of thalidomide in patients with inflammatory skin disease. Clin Exp Dermatol 2024; 49:168-170. [PMID: 37652049 DOI: 10.1093/ced/llad294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/06/2023] [Accepted: 09/21/2023] [Indexed: 09/02/2023]
Abstract
This retrospective review summarizes the effectiveness and safety concerns around using thalidomide in the treatment of inflammatory dermatoses.
Collapse
Affiliation(s)
- Sheila M McSweeney
- Department of Photodermatology, St John's Institute of Dermatology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Stephen Mounsey
- Department of Photodermatology, St John's Institute of Dermatology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Nisha Arujuna
- Department of Photodermatology, St John's Institute of Dermatology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Trish Garibaldinos
- Department of Photodermatology, St John's Institute of Dermatology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Robert Sarkany
- Department of Photodermatology, St John's Institute of Dermatology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| | - Hiva Fassihi
- Department of Photodermatology, St John's Institute of Dermatology, Guy's and St Thomas' Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
103
|
Gandhi SM, Paal E, Nylen ES. An Atypical Cause of Hoarseness in a Patient With Thyroid Nodules. Mil Med 2024; 189:e414-e416. [PMID: 37405706 DOI: 10.1093/milmed/usad234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/06/2023] [Accepted: 06/23/2023] [Indexed: 07/06/2023] Open
Abstract
Hoarseness due to vocal fold paresis (VFP) has a multitude of etiologies including systemic lupus erythematosus (SLE). During a clinical evaluation of a 58-year-old woman with long-standing hoarseness, an incidental finding of thyroid nodules was found to have VFP. Direct laryngoscopy and vocal fold biopsy confirmed the source was an inflammatory process involving the cricoarytenoid joint of the right hemilarynx. A presumptive diagnosis of SLE was made 3 years before meeting the clinical criteria of overt SLE. The VFP debut of SLE is extremely rare, and a literature review includes a handful of case reports (4 of a total of 37) since 1959. Only partial recovery of laryngeal function using glucocorticoids and Plaquenil was accomplished in the current case.
Collapse
Affiliation(s)
- Shruti M Gandhi
- Endocrinology Service, Veterans Affairs Medical Center, Washington, DC 20422, USA
| | - Edina Paal
- Pathology Department, Veterans Affairs Medical Center, Washington, DC 20422, USA
| | - Eric S Nylen
- Endocrinology Service, Veterans Affairs Medical Center, Washington, DC 20422, USA
| |
Collapse
|
104
|
Lee ARYB, Yau CE, Chua CKT, Cheng WL, Chia AJL, Wong SY, Kow NY, Gong L, Lee BTK, Ling LH, Mak A, Loh TP, Tay SH. High-density lipoprotein cholesterol subfraction HDL2 is associated with improved endothelial function in systemic lupus erythematosus. Lupus Sci Med 2024; 11:e001030. [PMID: 38262630 PMCID: PMC10806503 DOI: 10.1136/lupus-2023-001030] [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: 08/23/2023] [Accepted: 01/05/2024] [Indexed: 01/25/2024]
Abstract
OBJECTIVE Patients with systemic lupus erythematosus (SLE) have increased risk of premature atherosclerosis but the exact mechanisms remains unclear. Flow-mediated dilatation (FMD) is an established non-invasive assessment of vascular endothelial function. Lipoprotein subfractions may be better predictors of FMD than conventional cholesterol measurements. We tested the hypothesis that lipoprotein subfractions are independently associated with FMD. METHODS Forty-one consecutive adult patients with SLE without known cardiovascular risk factors or disease were recruited in this cross-sectional study. Endothelial function and early atherosclerosis were assessed by brachial FMD and common carotid artery (CCA) intima-media thickness (IMT). High-density lipoprotein (HDL)/low-density lipoprotein (LDL) subfractions were measured. Machine learning models were also constructed to predict FMD and CCA IMT. RESULTS Median FMD was 4.48% (IQR 5.00%) while median IMT was 0.54 mm (IQR 0.12 mm). Univariate analysis showed lower LDL1 (r=-0.313, p<0.05) and higher HDL2 subfractions (r=0.313, p<0.05) were significantly associated with higher log-transformed FMD. In a multiple linear regression model, HDL2 (β=0.024, SE=0.012, p<0.05) remained an independent predictor of higher FMD after adjusting for age, body mass index, LDL1 and systolic blood pressure. The machine learning model included parameters such as HDL2 (positive association), prednisolone dose, LDL cholesterol and LDL1 for prediction of FMD (r=0.433, p<0.01). Age, LDL cholesterol and systolic blood pressure were independently associated with higher CCA IMT after adjusting for body mass index and HDL2. CONCLUSIONS HDL 2, a large HDL particle, was independently associated with greater FMD and may be a biomarker of vascular health in SLE.
Collapse
Affiliation(s)
| | - Chun En Yau
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Cheryl Kai Ting Chua
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore
| | - Wan Ling Cheng
- Department of Laboratory Medicine, National University Hospital, Singapore
| | | | - Shi Yin Wong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nien Yee Kow
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lingli Gong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Bernett Teck Kwong Lee
- Centre for Biomedical Informatics, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Lieng Hsi Ling
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore
| | - Anselm Mak
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Hospital, Singapore
| | - Sen Hee Tay
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Rheumatology, Department of Medicine, National University Hospital, Singapore
| |
Collapse
|
105
|
Fan W, Wei B, Chen X, Zhang Y, Xiao P, Li K, Zhang YQ, Huang J, Leng L, Bucala R. Potential role of RhoA GTPase regulation in type interferon signaling in systemic lupus erythematosus. Arthritis Res Ther 2024; 26:31. [PMID: 38243295 PMCID: PMC10799493 DOI: 10.1186/s13075-024-03263-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by abnormal activation of the type I interferon (IFN) pathway, which results in tissue inflammation and organ damage. We explored the role of the RhoA GTPase in the type I IFN activation pathway to provide a potential basis for targeting GTPase signaling for the treatment of SLE. METHODS Total RNA was extracted from peripheral blood mononuclear cells (PBMCs) of SLE patients and healthy controls, and the mRNA expression levels of RhoA and IFN-stimulated genes were measured by SYBR Green quantitative reverse transcriptase-polymerase chain reaction. IFN-a-stimulated response element (ISRE)-luciferase reporter gene assays and Western blotting were conducted to assess the biologic function of RhoA. An enzyme-linked immunoassay (ELISA) measured C-X-C motif chemokine ligand 10 (CXCL10) protein expression. RESULTS Our studies demonstrate that the expression of RhoA in the PBMCs of SLE subjects was significantly higher than in healthy controls and positively correlated with type I IFN scores and type I IFN-stimulated gene (ISGs) expression levels. SiRNA-mediated knockdown of RhoA and the RhoA/ROCK inhibitor Y27632 reduced the activity of the type I IFN-induced ISRE, the signal transducer and activator of transcription 1 (STAT-1) phosphorylation, and the expression of CXCL10 and 2'-5'-oligoadenylate synthetase 1 (OAS1). Finally, we verified that Y27632 could significantly down-regulate the OAS1 and CXCL10 expression levels in the PBMCs of SLE patients. CONCLUSION Our study shows that RhoA positively regulates the activation of the type I IFN response pathway. Reducing the expression level of RhoA inhibits the abnormal activation of the type I IFN system, and the RhoA/ROCK inhibitor Y27632 decreases aberrant type I IFN signaling in SLE PBMCs, suggesting the possibility of targeting the RhoA GTPase for the treatment of SLE.
Collapse
Affiliation(s)
- Wei Fan
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China.
| | - Bo Wei
- Department of Rheumatology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Xuyan Chen
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Yi Zhang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Pingping Xiao
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Kaiyan Li
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Yi Qin Zhang
- Department of Nephrology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Jinmei Huang
- Department of Rheumatology and Immunology, the Second Affiliated Hospital of Xiamen Medical College, Xiamen Medical College, Xiamen, 361021, China
| | - Lin Leng
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Richard Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
106
|
Tanaka H, Okada Y, Nakayamada S, Miyazaki Y, Sonehara K, Namba S, Honda S, Shirai Y, Yamamoto K, Kubo S, Ikari K, Harigai M, Sonomoto K, Tanaka Y. Extracting immunological and clinical heterogeneity across autoimmune rheumatic diseases by cohort-wide immunophenotyping. Ann Rheum Dis 2024; 83:242-252. [PMID: 37903543 PMCID: PMC10850648 DOI: 10.1136/ard-2023-224537] [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: 06/09/2023] [Accepted: 09/13/2023] [Indexed: 11/01/2023]
Abstract
OBJECTIVE Extracting immunological and clinical heterogeneity across autoimmune rheumatic diseases (AIRDs) is essential towards personalised medicine. METHODS We conducted large-scale and cohort-wide immunophenotyping of 46 peripheral immune cells using Human Immunology Protocol of comprehensive 8-colour flow cytometric analysis. Dataset consisted of >1000 Japanese patients of 11 AIRDs with deep clinical information registered at the FLOW study, including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). In-depth clinical and immunological characterisation was conducted for the identified RA patient clusters, including associations of inborn human genetics represented by Polygenic Risk Score (PRS). RESULTS Multimodal clustering of immunophenotypes deciphered underlying disease-cell type network in immune cell, disease and patient cluster resolutions. This provided immune cell type specificity shared or distinct across AIRDs, such as close immunological network between mixed connective tissue disease and SLE. Individual patient-level clustering dissected patients with AIRD into several clusters with different immunological features. Of these, RA-like or SLE-like clusters were exclusively dominant, showing immunological differentiation between RA and SLE across AIRDs. In-depth clinical analysis of RA revealed that such patient clusters differentially defined clinical heterogeneity in disease activity and treatment responses, such as treatment resistance in patients with RA with SLE-like immunophenotypes. PRS based on RA case-control and within-case stratified genome-wide association studies were associated with clinical and immunological characteristics. This pointed immune cell type implicated in disease biology such as dendritic cells for RA-interstitial lung disease. CONCLUSION Cohort-wide and cross-disease immunophenotyping elucidate clinically heterogeneous patient subtypes existing within single disease in immune cell type-specific manner.
Collapse
Affiliation(s)
- Hiroaki Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
- Department of Statistical Genetics, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| | - Shingo Nakayamada
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Yusuke Miyazaki
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka, Japan
| | - Suguru Honda
- Department of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University School of Medicine Graduate School of Medicine, Suita, Osaka, Japan
- Laboratory of Children's health and Genetics, Division of Health Science, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Kubo
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Katsunori Ikari
- Department of Orthopedics, Tokyo Women's Medical University, Tokyo, Japan
| | - Masayoshi Harigai
- Department of Rheumatology, Department of Internal Medicine, Tokyo Women's Medical University, Shinjuku-ku, Tokyo, Japan
| | - Koshiro Sonomoto
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
- Department of Clinical Nursing, School of Health Sciences, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
107
|
Alduraibi FK, Tsokos GC. Lupus Nephritis Biomarkers: A Critical Review. Int J Mol Sci 2024; 25:805. [PMID: 38255879 PMCID: PMC10815779 DOI: 10.3390/ijms25020805] [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: 12/06/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Lupus nephritis (LN), a major complication in individuals diagnosed with systemic lupus erythematosus, substantially increases morbidity and mortality. Despite marked improvements in the survival of patients with severe LN over the past 50 years, complete clinical remission after immunosuppressive therapy is achieved in only half of the patients. Therefore, timely detection of LN is vital for initiating prompt therapeutic interventions and improving patient outcomes. Biomarkers have emerged as valuable tools for LN detection and monitoring; however, the complex role of these biomarkers in LN pathogenesis remains unclear. Renal biopsy remains the gold standard for the identification of the histological phenotypes of LN and guides disease management. However, the molecular pathophysiology of specific renal lesions remains poorly understood. In this review, we provide a critical, up-to-date overview of the latest developments in the field of LN biomarkers.
Collapse
Affiliation(s)
- Fatima K. Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Medicine, Division of Clinical Immunology and Rheumatology, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - George C. Tsokos
- Department of Medicine, Division of Clinical Immunology and Rheumatology, Beth Israel Deaconess Medical Center, Harvard Teaching Hospital, Boston, MA 02215, USA
| |
Collapse
|
108
|
Liu A, Li Z, Zeng J, Peng Y, Wang S, Bi X, Zhao Z, Zhou S, Zhao AZ, Mu Y, Li F. ω-3 polyunsaturated fatty acid alleviates systemic lupus erythematosus by suppressing autoimmunity in a murine model. Int Immunopharmacol 2024; 126:111299. [PMID: 38043268 DOI: 10.1016/j.intimp.2023.111299] [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: 08/24/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune inflammatory disease that damages multiple organs by the production of autoantibodies. Numerous research studies have demonstrated the anti-inflammatory effects of ω-3 polyunsaturated fatty acids (PUFAs). A diet rich in ω-3 PUFAs reduces chronic inflammatory and autoimmune conditions. Herein, we investigated the protective effect of ω-3 PUFAs against autoimmune injury in SLE. In a TMPD-induced mouse model of SLE, supplementation with eicosapentaenoic acid (EPA)-rich (97%) fish oil was found to alleviate systemic autoimmune phenotypes such as ascites, lipogranulomas and serum dsDNA levels. In addition, EPA also significantly improved renal manifestations, reducing proteinuria, glomerulonephritis, and immune complex deposition. Mechanistically, ω-3 PUFAs were shown to modulate the differentiation of B lymphocyte subsets of primary splenic lymphocytes in the spontaneous murine lupus model MRL/MpJ-Faslpr in vitro, specifically that both EPA and DHA suppressed the number of total B cells, B1B2 cells and plasma cells. Concurrently, they were also found to promote the secretion of the anti-inflammatory cytokine IL10, mainly produced by Breg and Treg cells. Thus, nutritional supplementation with ω-3 PUFAs can regulate B cell's differentiation and anti-inflammatory function and strongly prevent autoimmune responses and lupus nephritis. The diets balance between ω-6 and ω-3 PUFAs intake may represent a promising treatment strategy to prevent or delay the onset of SLE.
Collapse
Affiliation(s)
- Aolu Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhuang Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jingwen Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yuerong Peng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shuai Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinyun Bi
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhenggang Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Sujin Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Allan Zijian Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Yunping Mu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
109
|
Li X, Zhang Y, Li B, Li J, Qiu Y, Zhu Z, Hua H. An immunomodulatory antibody-drug conjugate targeting BDCA2 strongly suppresses plasmacytoid dendritic cell function and glucocorticoid responsive genes. Rheumatology (Oxford) 2024; 63:242-250. [PMID: 37184875 DOI: 10.1093/rheumatology/kead219] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/02/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023] Open
Abstract
OBJECTIVES Blood dendritic cell antigen 2 (BDCA2) is exclusively expressed on plasmacytoid dendritic cells (pDCs) whose uncontrolled production of type I IFN (IFN-I) is crucial in pathogenesis of SLE and other autoimmune diseases. Although anti-BDCA2 antibody therapy reduced disease activity in SLE patients, its clinical efficacy needs further improvement. We developed a novel glucocorticoid receptor agonist and used it as a payload to conjugate with an anti-BDCA2 antibody to form an BDCA2 antibody-drug conjugate (BDCA2-ADC). The activation of BDCA2-ADC was evaluated in vitro. METHODS Inhibitory activity of BDCA2-ADC was evaluated in peripheral blood mononuclear cells or in purified pDCs under ex vivo toll-like receptor agonistic stimulation. The global gene regulation in purified pDCs was analysed by RNA-seq. The antigen-dependent payload delivery was measured by reporter assay. RESULTS The BDCA2-ADC molecule causes total suppression of IFNα production and broader inhibition of inflammatory cytokine production compared with the parental antibody in human pDCs. Global gene expression analysis confirmed that the payload and antibody acted synergistically to regulate both type I IFN signature genes and glucocorticoid responsive genes in pDCs. CONCLUSION Taken together, these data suggest dual mechanisms of BDCA2-ADC on pDCs and the potential for BDCA2-ADC to be the first ADC treatment for SLE in the world and a better treatment option than anti-BDCA2 antibody for SLE patients.
Collapse
Affiliation(s)
- Xi Li
- Duality Biologics, Ltd, Shanghai, P.R. China
| | - Yu Zhang
- Duality Biologics, Ltd, Shanghai, P.R. China
| | - Bing Li
- Duality Biologics, Ltd, Shanghai, P.R. China
| | - Jian Li
- Duality Biologics, Ltd, Shanghai, P.R. China
| | - Yang Qiu
- Duality Biologics, Ltd, Shanghai, P.R. China
| | | | - Haiqing Hua
- Duality Biologics, Ltd, Shanghai, P.R. China
| |
Collapse
|
110
|
Suh CH, Lee Y, Yoo SB, Quasny H, Navarro Rojas AA, Hammer A, Song YW, Kang YM, Cho CS, Park W, Kwok SK, Lee SG, Chung WT, Bae SC. Efficacy and safety of intravenous belimumab in a subgroup of South Korean patients with systemic lupus erythematosus enrolled into a Phase 3, randomized, placebo-controlled trial in North East Asia. Int J Rheum Dis 2024; 27:e14997. [PMID: 38140854 DOI: 10.1111/1756-185x.14997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 10/23/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023]
Abstract
AIM This post hoc analysis evaluated the efficacy and safety of intravenous belimumab 10 mg/kg in the South Korean subgroup of patients with systemic lupus erythematosus (SLE) enrolled in the North East Asia (NEA) study (GSK Study BEL113750; NCT01345253). METHODS NEA was a double-blind, placebo-controlled, randomized Phase 3 trial. Patients with active, autoantibody-positive SLE were randomized 2:1 to belimumab or placebo plus standard therapy administered on Days 0, 14, and 28, and then every 28 days up to Week 48. The primary efficacy endpoint in this analysis was SLE Responder Index 4 (SRI-4) response rate at Week 52, defined as the proportion of patients achieving a ≥4-point reduction in Safety of Estrogens in Lupus Erythematosus National Assessment-SLE Disease Activity Index (SELENA-SLEDAI) score, no worsening (<0.3 increase from baseline) in Physician Global Assessment, no new British Isles Lupus Assessment Group (BILAG) A domain and <2 new BILAG B domain scores. RESULTS Among 100 South Korean patients enrolled in NEA, 54/66 (81.8%) belimumab- and 24/34 (70.6%) placebo-treated patients completed the double-blind phase. Significantly more belimumab- than placebo-treated patients achieved SRI-4 response at Week 52 (n = 35/66, 53.0% vs. n = 8/34, 23.5%; odds ratio [OR; 95% confidence interval (CI)]: 3.67 [1.45, 9.28]; p = .0061). The proportion of patients experiencing ≥1 adverse event was similar between groups (belimumab: n = 60/66, 90.9% vs. placebo: n = 31/34, 91.2%). No new safety signals emerged in this subgroup analysis. CONCLUSION Belimumab was efficacious for the treatment of SLE and well tolerated among the South Korean subgroup of patients from the NEA study.
Collapse
Affiliation(s)
- Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | | | | | - Holly Quasny
- Clinical Sciences, GSK, Durham, North Carolina, USA
| | | | - Anne Hammer
- Immunology Biostatistics, GSK, Collegeville, Pennsylvania, USA
| | - Yeong-Wook Song
- Division of Rheumatology, Seoul National University Hospital, Seoul, Korea
- Medical Research Center, Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea
| | - Young Mo Kang
- Division of Rheumatology, Department of Internal Medicine, Kyungpook National University Hospital, Daegu, Korea
| | - Chul-Soo Cho
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Won Park
- Medicine/Rheumatology, School of Medicine, IN-HA University, Incheon, Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Geun Lee
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Won Tae Chung
- Division of Rheumatology, Department of Internal Medicine, Dong-A University Hospital, Busan, Korea
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Hanyang University Institute for Rheumatology and Hanyang University Institute of Bioscience and Biotechnology, Seoul, Korea
| |
Collapse
|
111
|
Bao D, Drenkard C, Dunlop-Thomas C, Bayakly R, Lim SS. Direct medical charges in a population-based systemic lupus erythematosus cohort. J Med Econ 2024; 27:982-990. [PMID: 39049746 DOI: 10.1080/13696998.2024.2383047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/21/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
AIM This study aimed to obtain estimates for the direct medical charges associated with hospitalizations and emergency department visits of validated SLE cases in a diverse Systemic Lupus Erythematosus (SLE) population. METHODS The Georgians Organized Against Lupus (GOAL) cohort is a population-based cohort of adult SLE patients from metropolitan Atlanta, GA USA, an area having a diverse SLE population. The GOAL cohort aims to study the impact of social determinants of health (SDoH) on outcomes relevant to patients, healthcare providers, and policymakers. For this study, survey data collected during 2011-2012 was linked to the Georgia Hospital Discharge Database (HDD) to capture hospital admissions (HAs) and emergency department visits (EDVs) throughout Georgia from 2012 through 2013. Direct medical charges were summarized by HCU type among all patients, among those with actual visits, and by socio-demographics and healthcare factors. RESULTS Among 829 patients (94% women, 78% Black, 64% non-private insurance, 64% not-employed, mean age of 46), 170 (20.5%) and 300 (36.2%) participants had at least one HA and one EDV in 1-year of follow-up, respectively, with 111(13.4%) having both HA and EDV. On average, each patient experienced 0.38 HAs and 0.91 EDVs, with per-patient direct medical charges of $14,968 for HAs & $3,022 for EDVs, and $39,645 per HA & $3,305 per EDV. Patients with higher social vulnerability or more severe disease had higher charges for both HA and EDV (p < 0.01), likely due to the delayed care and neglected health needs leading to more advanced and costly medical treatments. Living below the federal poverty level was associated with higher charges for EDVs (p < 0.001) but with lower charges for HAs (p = 0.036). CONCLUSIONS This study underscores the economic burden of SLE on vulnerable populations, emphasizing the importance of including socio-economic factors in healthcare planning. Policy efforts should prioritize reducing disparities in access to care and implementing preventive strategies.
Collapse
Affiliation(s)
| | - Cristina Drenkard
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, GA, USA
| | - Charmayne Dunlop-Thomas
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rana Bayakly
- Georgia Department of Public Health, Atlanta, GA, USA
| | - S Sam Lim
- Department of Medicine, Division of Rheumatology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
112
|
Wu Y, Weng C, Zhou Y, Zhu Q, Liu Y, Zheng J, Yang B, Cao W, Yuan L, Yang M, Deng D. A comprehensive exploration of the heterogeneity of immune cells in Han and Zang systemic lupus erythematosus patients via single-cell RNA sequencing. Genomics 2024; 116:110770. [PMID: 38128704 DOI: 10.1016/j.ygeno.2023.110770] [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: 07/02/2023] [Revised: 11/16/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Systemic Lupus Erythematosus (SLE) is an autoimmune sickness with unclear pathogenesis. The goal of this research was to reveal the heterogeneity of immune cells in SLE patients of Han and Zang nationality by single-cell RNA sequencing (scRNA-seq) and bioinformatics profiling. METHODS A total of 94,102 peripheral blood mononuclear cells (PBMCs) from six volunteers with SLE (3 Zang, 3 Han) and six healthy controls were first conducted through scRNA-seq analysis. The immune cell subsets in the pathogenesis of SLE were analyzed as well. Real-time quantitative PCR (RT-qPCR) was applied to confirm the results of sc-RNA seq analysis. RESULTS For the Tibetan samples, the ratios of Naïve CD4 RPS4Y1 cells, Naïve CD4 cells, Memory BC CD24 and Memory BC differed significantly between the SLE and control samples, while that of CD8 CTL MAL cells was significantly different between the two groups in Han nationality samples. Variable differentiation states of CD8 CTL MAL cells, CD8 CTL GZMK cells, and Naïve CD4 cells were detected through pseudotime analysis. Moreover, T-cell receptor (TCR) abundance was notably higher in Tibetan SLE specimens than that in controls, while B-cell receptor (BCR) abundance in Tibetan and Han samples was higher than in control groups. CONCLUSIONS In summary, the immune cellular heterogeneity of SLE patients both Han and Zang nationality was explored based on various bioinformatics approaches, providing new perspectives for immunological characteristics of SLE among different ethnic groups.
Collapse
Affiliation(s)
- Yongzhuo Wu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Chongjun Weng
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, PR China
| | - Yali Zhou
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Qinghuan Zhu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Yingying Liu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Junjuan Zheng
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650201, PR China
| | - Binbin Yang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Wenting Cao
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Limei Yuan
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China
| | - Meng Yang
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China; Department of Dermatology, The Third Affiliated Hospital of Guangxi Medical University, Nanning 530031, PR China
| | - Danqi Deng
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, PR China.
| |
Collapse
|
113
|
Pandey SP, Bhaskar R, Han SS, Narayanan KB. Autoimmune Responses and Therapeutic Interventions for Systemic Lupus Erythematosus: A Comprehensive Review. Endocr Metab Immune Disord Drug Targets 2024; 24:499-518. [PMID: 37718519 DOI: 10.2174/1871530323666230915112642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/05/2023] [Accepted: 07/22/2023] [Indexed: 09/19/2023]
Abstract
Systemic Lupus Erythematosus (SLE) or Lupus is a multifactorial autoimmune disease of multiorgan malfunctioning of extremely heterogeneous and unclear etiology that affects multiple organs and physiological systems. Some racial groups and women of childbearing age are more susceptible to SLE pathogenesis. Impressive progress has been made towards a better understanding of different immune components contributing to SLE pathogenesis. Recent investigations have uncovered the detailed mechanisms of inflammatory responses and organ damage. Various environmental factors, pathogens, and toxicants, including ultraviolet light, drugs, viral pathogens, gut microbiome metabolites, and sex hormones trigger the onset of SLE pathogenesis in genetically susceptible individuals and result in the disruption of immune homeostasis of cytokines, macrophages, T cells, and B cells. Diagnosis and clinical investigations of SLE remain challenging due to its clinical heterogeneity and hitherto only a few approved antimalarials, glucocorticoids, immunosuppressants, and some nonsteroidal anti-inflammatory drugs (NSAIDs) are available for treatment. However, the adverse effects of renal and neuropsychiatric lupus and late diagnosis make therapy challenging. Additionally, SLE is also linked to an increased risk of cardiovascular diseases due to inflammatory responses and the risk of infection from immunosuppressive treatment. Due to the diversity of symptoms and treatment-resistant diseases, SLE management remains a challenging issue. Nevertheless, the use of next-generation therapeutics with stem cell and gene therapy may bring better outcomes to SLE treatment in the future. This review highlights the autoimmune responses as well as potential therapeutic interventions for SLE particularly focusing on the recent therapeutic advancements and challenges.
Collapse
Affiliation(s)
- Surya Prakash Pandey
- Aarogya Institute of Healthcare and Research, Jaipur, Rajasthan, 302033, India
- Department of Zoology, School of Science, IFTM University, Moradabad, Uttar Pradesh, 244102, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| |
Collapse
|
114
|
Jiang P, Yao C, Guo DA. Traditional Chinese medicine for the treatment of immune-related nephropathy: A review. Acta Pharm Sin B 2024; 14:38-66. [PMID: 38239236 PMCID: PMC10793104 DOI: 10.1016/j.apsb.2023.11.006] [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: 03/23/2023] [Revised: 09/18/2023] [Accepted: 10/24/2023] [Indexed: 01/22/2024] Open
Abstract
Immune-related nephropathy (IRN) refers to immune-response-mediated glomerulonephritis and is the main cause of end-stage renal failure. The pathogenesis of IRN is not fully understood; therefore, treatment is challenging. Traditional Chinese medicines (TCMs) have potent clinical effects in the treatment of the IRN conditions immunoglobulin A nephropathy, lupus nephropathy, and diabetic nephropathy. The underlying mechanisms mainly include its inhibition of inflammation; improvements to renal interstitial fibrosis, oxidative stress, autophagy, apoptosis; and regulation of immunity. In this review, we summarize the clinical symptoms of the three IRN subtypes and the use of TCM prescriptions, herbs, and bioactive compounds in treating IRN, as well as the potential mechanisms, intending to provide a reference for the future study of TCM as IRN treatments.
Collapse
Affiliation(s)
- Pu Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Changliang Yao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - De-an Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
115
|
Yang F, Zhang Y, Dong L, Song Z. Cordyceps cicadae ameliorates inflammatory responses, oxidative stress, and fibrosis by targeting the PI3K/mTOR-mediated autophagy pathway in the renal of MRL/lpr mice. Immun Inflamm Dis 2024; 12:e1168. [PMID: 38270299 PMCID: PMC10808846 DOI: 10.1002/iid3.1168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND The vast majority of systemic lupus erythematosus patients develop lupus nephritis (LN) with severe renal manifestations, such as inflammatory responses, oxidative stress, and fibrosis. The purpose of this research was to investigate Cordyceps cicadae as a potential therapeutic target for treating inflammatory responses, oxidative stress, and fibrosis in LN. METHODS The effects of C. cicadae on lupus symptoms in mice with LN were determined. MRL/lpr mice were treated with C. cicadae (4 g/kg/day, i.e., CC group, n = 8) or an equal volume of saline (model group, n = 8), and MRL/MP mice were treated with an equal volume of saline (control group, n = 8). Renal function indices, renal pathology, inflammatory markers, oxidative stress markers, and renal interstitial fibrosis levels were evaluated after C. cicadae treatment. Western blot analysis was performed to investigate the effect of C. cicadae on the expression of fibrosis biomarkers and the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR)-mediated autophagy pathway in the renal tissues of mice. RESULTS C. cicadae ameliorated renal lesions, the inflammatory response, and oxidative stress damage in MRL/lpr mice. C. cicadae treatment inhibited renal fibrosis (16.31 ± 4.16 vs. 31.25 ± 5.61) and downregulated the expression of the fibrosis biomarkers alpha-smooth muscle actin, fibronectin, and collagen I (COL I) in the kidneys of MRL/lpr mice. In addition, further research showed that the PI3K/mTOR-mediated autophagy pathway was involved in C. cicadae-mediated effects on renal fibrosis in MRL/lpr mice. Furthermore, the therapeutic effect of C. cicadae on repairing renal fibrosis and damage in MRL/lpr mice was abolished by the PI3K agonist 740 Y-P. CONCLUSIONS The findings of the present research showed that C. cicadae could alleviate inflammatory responses, oxidative stress, and fibrosis in the renal tissues of mice with LN by targeting the PI3K/mTOR-mediated autophagy pathway.
Collapse
Affiliation(s)
- Feng Yang
- Department of RheumatologyYantai Hospital of Traditional Chinese MedicineYantai CityShandongChina
| | - Yanyan Zhang
- Department of RheumatologyYantai Hospital of Traditional Chinese MedicineYantai CityShandongChina
| | - Lei Dong
- Department of RheumatologyYantai Hospital of Traditional Chinese MedicineYantai CityShandongChina
| | - Zhichao Song
- Department of RheumatologyYantai Hospital of Traditional Chinese MedicineYantai CityShandongChina
| |
Collapse
|
116
|
Ding H, Shen Y, Hong SM, Xiang C, Shen N. Biomarkers for systemic lupus erythematosus - a focus on organ damage. Expert Rev Clin Immunol 2024; 20:39-58. [PMID: 37712757 DOI: 10.1080/1744666x.2023.2260098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/16/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is complex autoimmune disease with heterogenous manifestations, unpredictable disease course and response to treatment. One of the critical needs in SLE management is the identification of reliable biomarkers that can aid in early diagnosis, accurate monitoring of disease activity, and assessment of treatment response. AREAS COVERED In the current review, we focus on the commonly affected organs (skin, kidney, and nervous system) in SLE to summarize the emerging biomarkers that show promise in disease diagnosis, monitoring and treatment response assessment. The subtitles within each organ domain were determined based on the most relevant and promising biomarkers for that specific organ damage. EXPERT OPINION Biomarkers have the potential to significantly benefit the management of SLE by aiding in diagnosis, disease activity monitoring, prognosis, and treatment response assessment. However, despite decades of research, none has been validated and implemented for routine clinical use. Novel biomarkers could lead to the development of precision medicine for SLE, guide personalized treatment, and improve patient outcomes. Challenges in biomarker research in SLE include defining clear and clinically relevant questions, accounting for the heterogeneity of SLE, and confirming initial findings in larger, multi-center, multi-ethnic, independent cohorts that reflect real-world clinical scenarios.
Collapse
Affiliation(s)
- Huihua Ding
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Yiwei Shen
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Soon-Min Hong
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Chunyan Xiang
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | - Nan Shen
- Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- China-Australia Centre for Personalized Immunology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Department of Rheumatology, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
- Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Collaborative Innovation Centre for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
117
|
Bohat R, Liang X, Chen Y, Xu C, Zheng N, Guerrero A, Hou J, Jaffery R, Egan NA, Li Y, Tang Y, Unsal E, Robles A, Chen S, Major AM, Elldakli H, Chung SH, Liang H, Hicks MJ, Du Y, Lin JS, Chen X, Mohan C, Peng W. Fas lpr gene dosage tunes the extent of lymphoproliferation and T cell differentiation in lupus. Clin Immunol 2024; 258:109874. [PMID: 38113962 DOI: 10.1016/j.clim.2023.109874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/23/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
Sle1 and Faslpr are two lupus susceptibility loci that lead to manifestations of systemic lupus erythematosus. To evaluate the dosage effects of Faslpr in determining cellular and serological phenotypes associated with lupus, we developed a new C57BL/6 (B6) congenic lupus strain, B6.Sle1/Sle1.Faslpr/+ (Sle1homo.lprhet) and compared it with B6.Faslpr/lpr (lprhomo), B6.Sle1/Sle1 (Sle1homo), and B6.Sle1/Sle1.Faslpr/lpr (Sle1homo.lprhomo) strains. Whereas Sle1homo.lprhomo mice exhibited profound lymphoproliferation and early mortality, Sle1homo.lprhet mice had a lifespan comparable to B6 mice, with no evidence of splenomegaly or lymphadenopathy. Compared to B6 monogenic lupus strains, Sle1homo.lprhet mice exhibited significantly elevated serum ANA antibodies and increased proteinuria. Additionally, Sle1homo.lprhet T cells had an increased propensity to differentiate into Th1 cells. Gene dose effects of Faslpr were noted in upregulating serum IL-1⍺, IL-2, and IL-27. Taken together, Sle1homo.lprhet strain is a new C57BL/6-based model of lupus, ideal for genetic studies, autoantibody repertoire investigation, and for exploring Th1 effector cell skewing without early-age lymphoproliferative autoimmunity.
Collapse
Affiliation(s)
- Ritu Bohat
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Xiaofang Liang
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Yanping Chen
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, United States of America
| | - Chunyu Xu
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Ningbo Zheng
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Ashley Guerrero
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Jiakai Hou
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Roshni Jaffery
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Nicholas A Egan
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Yaxi Li
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, United States of America
| | - Yitao Tang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America; UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson, Houston, TX 77030, United States of America
| | - Esra Unsal
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Adolfo Robles
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Si Chen
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Angela M Major
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, United States of America
| | - Hadil Elldakli
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Sang-Hyuk Chung
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America; Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - M John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, United States of America
| | - Yong Du
- Department of Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, United States of America
| | - Jamie S Lin
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States of America
| | - Xiqun Chen
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, United States of America; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, United States of America; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, United States of America; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, United States of America.
| |
Collapse
|
118
|
Parodis I, Depascale R, Doria A, Anders HJ. When should targeted therapies be used in the treatment of lupus nephritis: Early in the disease course or in refractory patients? Autoimmun Rev 2024; 23:103418. [PMID: 37625673 DOI: 10.1016/j.autrev.2023.103418] [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: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Although the prognosis of lupus nephritis (LN) has improved over the last few decades, 5-20% of patients still progress to kidney failure. Hence, there is an unmet need to improve the management of LN. Two novel drugs, belimumab and voclosporin, have been recently approved for LN and obinutuzumab is in the late stage of development. In randomised controlled trials (RCTs), all these drugs, added to the standard-of-care, were more effective than standard-of-care alone in achieving renal response. Now the question is: should these new drugs be used early in the disease course or just in refractory patients? The main reasons supporting the early use are based on the RCTs that demonstrated benefits when combinatory regimen was initiated early in incident and relapsing patients leading to a higher proportion of patients to achieve renal response, hence reducing nephron loss and the risk of kidney failure. The main reasons supporting the use of the combinatory regimens primarily in relapsing/refractory patients acknowledge that many patients responded well even without add-on medications, allowing a more economic use of innovative and costly drugs. However, good predictors of renal response to standard-of-care are lacking and, thus, the decision of adding new treatments early or just in refractory or relapsing patients has to consider drug access, risks of over or undertreatment, and preservation of kidney function in high-risk individuals.
Collapse
Affiliation(s)
- Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Gastroenterology, Dermatology, and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Roberto Depascale
- Deparment of Medicine DIMED, Division of Rheumatology, University of Padua, Padua, Italy
| | - Andrea Doria
- Deparment of Medicine DIMED, Division of Rheumatology, University of Padua, Padua, Italy.
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Hospital of the Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
119
|
Liu S, Si S, Li J, Zhao Y, Yu Q, Xue F. Association between type 1 diabetes and systemic lupus erythematosus: a Mendelian randomization study. Clin Rheumatol 2024; 43:41-48. [PMID: 37947970 DOI: 10.1007/s10067-023-06800-8] [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: 03/18/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVES Observational studies have shown that there is a bidirectional relationship between type 1 diabetes (T1D) and systemic lupus erythematosus (SLE); the causality of this association remains elusive and may be affected by confusion and reverse causality. There is also a lack of large-scale randomized controlled trials to verify. Therefore, this Mendelian randomization (MR) study aimed to investigate the causal association between T1D and SLE. METHODS We aggregated data using publicly available genome-wide association studies (GWAS), all from European populations. Select independent (R2 < 0.001) and closely related to exposure (P < 5 × 10-8) as instrumental variables (IVs). The inverse-variance weighted (IVW) method was used as the primary method. We also used MR-Egger, the weighted median method, MR-Robust, MR-Lasso, and other methods leveraged as supplements. RESULTS T1D had a positive causal association with SLE (IVW, odds ratio [OR] = 1.358, 95% confidence interval [CI], 1.205 - 1.530; P < 0.001). The causal association was verified in an independent validation set (IVW, OR = 1.137, 95% CI, 1.033 - 1.251; P = 0.001). SLE had a positive causal association with T1D (IVW, OR = 1.108, 95% CI, 1.074 - 1.144; P < 0.001). The causal association was verified in an independent validation set (IVW, OR = 1.085, 95% CI, 1.046 - 1.127; P < 0.001). These results have also been verified by sensitivity analysis. CONCLUSION The MR analysis results indicated a causal association between T1D and SLE. Therefore, further research is needed to clarify the potential biological mechanism between T1D and SLE. Key Points • Observational studies have shown that there is a bidirectional relationship between T1D and SLE. • We evaluated causal effects between T1D and SLE by Mendelian randomization analyses. • The MR analysis results indicated a causal association between T1D and SLE.
Collapse
Affiliation(s)
- Shulin Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Culture West Road, Jinan, 250012, Shandong, China
- Healthcare Big Data Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Shucheng Si
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Culture West Road, Jinan, 250012, Shandong, China
- Healthcare Big Data Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Jiqing Li
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Culture West Road, Jinan, 250012, Shandong, China
- Healthcare Big Data Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yingqi Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Culture West Road, Jinan, 250012, Shandong, China
- Healthcare Big Data Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Qingqing Yu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Culture West Road, Jinan, 250012, Shandong, China
- Healthcare Big Data Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Culture West Road, Jinan, 250012, Shandong, China.
- Healthcare Big Data Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
120
|
Panda AK, Ranjan S, Sahu JK. Efficacy of baricitinib for the treatment of systemic lupus erythematosus patients: A meta-analysis of randomized controlled trials. Int J Rheum Dis 2024; 27:e14964. [PMID: 37950554 DOI: 10.1111/1756-185x.14964] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/10/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by abnormal autoantibody production, inflammation, and organ damage. Most SLE treatment strategies aim to induce remission or reduce disease activity while avoiding flares. Baricitinib has been used effectively to manage various inflammatory diseases, and some randomized controlled trials (RCT) have shown that it is beneficial in treating SLE. The current study aims to assess the efficacy of baricitinib in treating SLE patients. MATERIALS AND METHODS Various databases such as PubMed, Scopus, and Science Direct were searched to obtain eligible studies for the present meta-analysis. Data such as baseline characteristics of patients, doses of the baricitinib, follow-up duration, and treatment outcome in the form of SLE responder index-4 (SRI-4) and lupus low disease activity state (LLDAS) were extracted. Combined odds ratio, 95% confidence interval, and probability values were calculated to study the efficacy of baricitinib in treating SLE patients. A p-value less than .05 was taken as significant. Comprehensive meta-analysis v3 was used for all analyses. RESULTS Three articles were found eligible for the present meta-analysis comprising 614 patients with placebo, 614 SLE patients receiving 4 mg, and 621 patients with 2 mg of baricitinib. Meta-analysis revealed a beneficial effect of 4 mg baricitinib in SLE patients compared to placebo, as measured by an increase in the SRI-4 (p = .006, OR = 1.370) and LLDAS (p = .083, OR = 1.252) rates. In contrast to the placebo group, however, patients receiving 2 mg of baricitinib exhibited no significant improvement. The trial sequential analysis revealed the need for additional RCTs to determine the role of baricitinib in treating SLE patients. CONCLUSION In treating SLE patients, administrating a higher dose of baricitinib (4 mg) may be effective. However, additional RCTs in different populations with larger sample sizes are required to validate our findings.
Collapse
Affiliation(s)
- Aditya K Panda
- Department of Biotechnology, Berhampur University, Bhanja Bihar, Berhampur, Odisha, India
- Centre of Excellence on "Bioprospecting of Ethnopharmaceuticals of Southern Odisha" (CoE-BESO), Berhampur University, Bhanja Bihar, Berhampur, Odisha, India
| | - Shovit Ranjan
- University Department of Zoology, Kolhan University, Chaibasa, Jharkhand, India
| | - Jayanta K Sahu
- Department of Biology, Odisha Adarsha Vidyalaya Kursud, Balangir, Odisha, India
| |
Collapse
|
121
|
Li Y, Wu Y, Huang J, Cao X, An Q, Peng Y, Zhao Y, Luo Y. A variety of death modes of neutrophils and their role in the etiology of autoimmune diseases. Immunol Rev 2024; 321:280-299. [PMID: 37850797 DOI: 10.1111/imr.13284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Neutrophils are important in the context of innate immunity and actively contribute to the progression of diverse autoimmune disorders. Distinct death mechanisms of neutrophils may exhibit specific and pivotal roles in autoimmune diseases and disease pathogenesis through the orchestration of immune homeostasis, the facilitation of autoantibody production, the induction of tissue and organ damage, and the incitement of pathological alterations. In recent years, more studies have provided in-depth examination of various neutrophil death modes, revealing nuances that challenge conventional understanding and underscoring their potential clinical utility in diagnosis and treatment. This review explores the multifaceted processes and characteristics of neutrophil death, with a focus on tailored investigations within various autoimmune diseases. It also highlights the potential interplay between neutrophil death and the landscape of autoimmune disorders. The review encapsulates the pertinent pathways implicated in various neutrophil death mechanisms across diverse autoimmune diseases while also charts possible avenues for future research.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yinlan Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingang Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xue Cao
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Qiyuan An
- School of Inspection and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yun Peng
- Department of Rheumatology and Clinical Immunology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Yi Zhao
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
122
|
Inês LS, Fredi M, Jesus D, Shumilova A, Franceschini F, Vital EM. What is the best instrument to measure disease activity in SLE? - SLE-DAS vs Easy BILAG. Autoimmun Rev 2024; 23:103428. [PMID: 37634675 DOI: 10.1016/j.autrev.2023.103428] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous condition making assessment of disease activity challenging. However, thorough assessment is essential to evaluate patients longitudinally, to guide therapeutic decisions, and for clinical trials. Currently, the most popular disease activity index in clinical practice and trials is SLEDAI-2K. Its main advantage is ease of use, but significant weaknesses of SLEDAI-2K are omission of several serious manifestations, inability to capture change within an organ system, and fixed severity weightings that are often inappropriate. Recently several groups have developing improved tools. We report here the debate held at CORA meeting on this issue. SLE-DAS includes 17 weighted clinical and laboratory parameters including continuous measures in 4 items with an online calculator. A higher sensitivity to change compared to SLEDAI-2K has been demonstrated in its validation studies. Easy BILAG is an improved format of the BILAG-2004 that retains its content but greatly simplified. Its scoring using a single-page form that incorporates concise definitions for key terms next to clinical items. Easy-BILAG demonstrates higher accuracy and less variability and increased and better user feedback compared to the standard BILAG-2004 format. Both the indices discussed at CORA showed an advantage in measuring disease activity compared to SLEDAI.
Collapse
Affiliation(s)
- Luís Sousa Inês
- Faculty of Health Sciences, Universidade da Beira Interior, Covilhã, Portugal; CHUC Lupus Clinic, Rheumatology Department, Centro Hospitalar e Universitário de Coimbra, Portugal
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit and Clinical and Experimental Science Department ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Diogo Jesus
- Faculty of Health Sciences, Universidade da Beira Interior, Covilhã, Portugal; Rheumatology Department, Centro Hospitalar de Leiria, Leiria, Portugal
| | - Anastasiia Shumilova
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom; V.A. Nasonova Research Institute of Rheumatology, Moscow, Russian Federation
| | - Franco Franceschini
- Rheumatology and Clinical Immunology Unit and Clinical and Experimental Science Department ASST Spedali Civili and University of Brescia, Brescia, Italy.
| | - Edward M Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
123
|
Miyake K, Shibata T, Fukui R, Murakami Y, Sato R, Hiranuma R. Endosomal Toll-Like Receptors as Therapeutic Targets for Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:97-108. [PMID: 38467975 DOI: 10.1007/978-981-99-9781-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Nucleic acid (NA)-sensing Toll-like receptors (TLRs) reside in the endosomal compartment of innate immune cells, such as macrophages and dendritic cells. NAs transported to the endosomal compartment are degraded by DNases and RNases. Degradation products, including single-stranded DNA, oligoRNA, and nucleosides, are recognized by TLR7, TLR8, and TLR9 to drive the defense responses against pathogens. NA degradation influences endosomal TLR responses by generating and degrading TLR ligands. TLR ligand accumulation because of impaired NA degradation causes constitutive TLR activation, leading to autoinflammatory and autoimmune diseases. Furthermore, some genes associated with these diseases promote endosomal TLR responses. Therefore, endosomal TLRs are promising therapeutic targets for TLR-mediated inflammatory diseases, and novel drugs targeting TLRs are being developed.
Collapse
Affiliation(s)
- Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Hiranuma
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
124
|
Barraclough M, Howe A, Soberanis A, Kakvan M, Chattu V, Bani‐Fatemi A, Engel L, Vitti M, Nalder E, Goverover Y, Gignac M, Bonilla D, Nielsen W, Anderson N, Tartaglia C, Nowrouzi‐Kia B, Touma Z. The Effects of Systemic Lupus-Related Cognitive Impairments on Activities of Daily Living and Life Role Participation: A Qualitative Framework Study. ACR Open Rheumatol 2024; 6:21-30. [PMID: 37964675 PMCID: PMC10789303 DOI: 10.1002/acr2.11624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/30/2023] [Indexed: 11/16/2023] Open
Abstract
OBJECTIVE Cognitive impairment (CI) in systemic lupus erythematosus (SLE) negatively impacts health-related quality of life leading to activity limitations. This qualitative study aimed to (1) explore the effect of SLE-related CI on activities of daily living and life role participation and (2) describe factors influencing activity restriction and life role participation. METHODS Semistructured, in-depth interviews of lived experience of CI in SLE were conducted with 24 participants with SLE. Sociodemographic and clinical data, and objective and subjective cognitive function, were collected to characterize participants. A qualitative thematic content analysis was undertaken guided by a framework analytical approach. RESULTS Participants reported problems in multiple cognitive domains, with multiple perceived causes. CI was felt to impact work, social, domestic, and family life, health, and independence. Five overarching themes were represented in the data: (1) characterization of SLE-reported CI, (2) perceived cause of CI, (3) perceived impact of CI on activities of daily living and life role participation, (4) adaptations for managing CI, and (5) influence of CI adaptations on activities of daily living and life role participation. CONCLUSION This study provides a better understanding of the patient experience of CI in SLE, how it impacts their lives, and what coping strategies they employ. It highlights the long-term challenges those with CI in SLE undergo and provides evidence for the urgent need to implement multidisciplinary treatment options. When managing CI, it may be beneficial to evaluate and understand available psychosocial support resources to help identify and reinforce relevant adaptations to improve health-related quality of life.
Collapse
Affiliation(s)
- Michelle Barraclough
- University Health Network, Toronto, Ontario, Canada, and The University of Manchester, NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, and Manchester Academic Health Science CentreManchesterUK
| | - Aaron Howe
- University of TorontoTorontoOntarioCanada
| | | | - Mahta Kakvan
- University Health Network, and University of Toronto Lupus Clinic, Toronto Western HospitalTorontoOntarioCanada
| | | | | | - Lisa Engel
- University of Manitoba, Winnipeg, Manitoba, and Institute for Work and HealthTorontoOntarioCanada
| | - Michelle Vitti
- University Health Network, and University of Toronto Lupus Clinic, Toronto Western HospitalTorontoOntarioCanada
| | | | | | - Monique Gignac
- University Health Network, University of Toronto, and Institute for Work and HealthTorontoOntarioCanada
| | - Dennisse Bonilla
- University Health Network, and University of Toronto Lupus Clinic, Toronto Western HospitalTorontoOntarioCanada
| | - Wils Nielsen
- University of Toronto Lupus Clinic and Western HospitalTorontoOntarioCanada
| | - Nicole Anderson
- University Health Network, and University of Toronto Lupus Clinic, Toronto Western HospitalTorontoOntarioCanada
| | | | | | - Zahi Touma
- University Health Network, and University of Toronto Lupus Clinic, Toronto Western HospitalTorontoOntarioCanada
| |
Collapse
|
125
|
París-Muñoz A, León-Triana O, Pérez-Martínez A, Barber DF. Helios as a Potential Biomarker in Systemic Lupus Erythematosus and New Therapies Based on Immunosuppressive Cells. Int J Mol Sci 2023; 25:452. [PMID: 38203623 PMCID: PMC10778776 DOI: 10.3390/ijms25010452] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The Helios protein (encoded by the IKZF2 gene) is a member of the Ikaros transcription family and it has recently been proposed as a promising biomarker for systemic lupus erythematosus (SLE) disease progression in both mouse models and patients. Helios is beginning to be studied extensively for its influence on the T regulatory (Treg) compartment, both CD4+ Tregs and KIR+/Ly49+ CD8+ Tregs, with alterations to the number and function of these cells correlated to the autoimmune phenomenon. This review analyzes the most recent research on Helios expression in relation to the main immune cell populations and its role in SLE immune homeostasis, specifically focusing on the interaction between T cells and tolerogenic dendritic cells (tolDCs). This information could be potentially useful in the design of new therapies, with a particular focus on transfer therapies using immunosuppressive cells. Finally, we will discuss the possibility of using nanotechnology for magnetic targeting to overcome some of the obstacles related to these therapeutic approaches.
Collapse
Affiliation(s)
- Andrés París-Muñoz
- Department of Immunology and Oncology and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain;
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; (O.L.-T.); (A.P.-M.)
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Odelaisy León-Triana
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; (O.L.-T.); (A.P.-M.)
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; (O.L.-T.); (A.P.-M.)
- IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Domingo F. Barber
- Department of Immunology and Oncology and NanoBiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain;
| |
Collapse
|
126
|
Chen F, Shi B, Liu W, Gong J, Gao J, Sun Y, Yang P. Circulating exosomal microRNAs as biomarkers of lupus nephritis. Front Immunol 2023; 14:1326836. [PMID: 38223506 PMCID: PMC10785001 DOI: 10.3389/fimmu.2023.1326836] [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: 10/24/2023] [Accepted: 12/08/2023] [Indexed: 01/16/2024] Open
Abstract
Objective Disruption in the delicate symphony of genes, microRNA (miRNA), or protein expression can result in the dysregulation of the immune system, leading to the devastating consequences such as lupus nephritis (LN). The capacity of exosomes to transport miRNAs between cells and modify the phenotype of recipient cells implies their involvement in persistent kidney inflammation. This study unveils identifying two previously undiscovered exosomal miRNAs in the serum of LN patients, offering potential solutions to the current challenges in LN diagnosis and management. Methods Initially, we used a reagent-based kit to isolate serum exosomes from patients with Systemic lupus erythematosus (SLE) and used Trizol method for total RNA extraction. Subsequently, we employed small RNA sequencing to screen for differential expression profiles of exosomal small RNAs. The RT-qPCR method was used to individually validate samples in both the screening and validation cohorts, enabling the identification of candidate small RNAs; specific to LN. We assessed the diagnostic potency using receiver operating characteristic (ROC) curve, and explored the biological roles of miRNAs using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Results Compared to SLE patients without LN, SLE patients accompanied by LN exhibited significantly spiked levels of exosomal hsa-miR-4796-5p and hsa-miR-7974. The duo of miRNAs, hsa-miR-4796-5p and hsa-miR-7974, exhibited promising potential as biomarkers for diagnosing LN, with an AUC exceeding 0.8. Correlation analysis revealed a strong positive association between these miRNAs and proteinuria, as well as the SLE Disease Activity Index (SLEDAI) score. Moreover, the levels of two miRNAs in LN patients were significantly elevated in comparison to other autoimmune nephritis conditions, such as immunoglobulin A nephropathy (IgAN) and diabetic nephropathy (DN). Furthermore, the bioinformatics analysis indicated that this miRNAs duo can play a pivotal role in the regulation of immune processes by modulating signal pathways, such as the mTOR and PI3K-Akt signaling pathway. Conclusion This study provides a new ground that serum exosomal miRNAs can effectively identify and predict LN in SLE patients.
Collapse
Affiliation(s)
- Fei Chen
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Bo Shi
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenjing Liu
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jianmin Gong
- College of Life Sciences, Yangtze University, Jingzhou, China
| | - Jia Gao
- Department of Orthopedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Sun
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Ping Yang
- Department of Clinical Laboratory, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
127
|
Arnaud L, Furie R, Morand EF, Aringer M, Peschken C, Desta B, Rapsomaniki E, Hedberg J, Knagenhjelm J, Seo C, Grünfeld Eén T, Sorrentino A, Tummala R, Stirnadel-Farrant HA, Ding B. Burden of systemic lupus erythematosus in clinical practice: baseline data from the SLE Prospective Observational Cohort Study (SPOCS) by interferon gene signature. Lupus Sci Med 2023; 10:e001032. [PMID: 38123459 DOI: 10.1136/lupus-2023-001032] [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: 08/23/2023] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE The longitudinal Systemic Lupus Erythematosus Prospective Observational Cohort Study (SPOCS) aims to assess SLE disease course overall and according to type I interferon 4 gene signature (IFNGS). Here, we describe SPOCS patient characteristics by IFNGS and baseline disease activity. METHODS SPOCS (NCT03189875) is an international study of patients with SLE according to Systemic Lupus International Collaborating Clinics (SLICC)/American College of Rheumatology (ACR) criteria. Enrolled patients from 135 centres in 8 countries were followed biannually for ≤3 years from June 2017 to November 2022. Baseline demographics, disease characteristics, organ system involvement/damage and flares were analysed descriptively according to SLE Disease Activity Index-2000 score (SLEDAI-2K <10/≥10) and IFNGS status (high/low). RESULTS The study population (n=823) was 93.2% female, with mean (SD) age 45.3 (13.9) years and 11.1 (9.2) years since diagnosis; 52.4% had baseline SLICC/ACR Damage Index score ≥1. Patients with SLEDAI-2K scores ≥10 (241 of 584, 41.3%) vs <10 were younger (mean 42.8 (13.7) vs 46.6 (14.2) years; nominal p=0.001), had shorter SLE duration (10.4 (8.6) vs 12.4 (9.6) years; nominal p=0.012) and more severe flares (12.9% vs 5.3%; nominal p=0.001). IFNGS-high patients (522 of 739, 70.6%) were younger than IFNGS-low patients at first SLE manifestation (30.0 (12.7) vs 36.8 (14.6) years; nominal p<0.001). Proportions of IFNGS-high patients differed according to race (nominal p<0.001), with higher proportions among Asian (83.3%) and black (86.5%) versus white patients (63.5%). Greater proportions of IFNGS-high versus IFNGS-low patients had haematological (12.6% vs 4.1%), immunological (74.4% vs 45.6%) or dermal (69.7% vs 62.2%) involvement. CONCLUSIONS We identified key characteristics of patients with high disease activity and/or elevated type I IFN signalling, populations with SLE with high unmet needs. Baseline SLEDAI-2K ≥10 was associated with shorter disease duration and more severe flares. IFNGS-high patients were younger at diagnosis and had distinct patterns of organ involvement, compared with IFNGS-low patients.
Collapse
Affiliation(s)
- Laurent Arnaud
- Department of Rheumatology, University Hospitals of Strasbourg and French National Reference Center for Rare Autoimmune Diseases (RESO), INSERM UMR-S 1109, Strasbourg, France
| | - Richard Furie
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York, USA
| | | | - Martin Aringer
- University Medical Center and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Christine Peschken
- Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Barnabas Desta
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | | | - Caroline Seo
- BioPharmaceuticals Medical, AstraZeneca, Gaithersburg, Maryland, USA
| | | | | | - Raj Tummala
- BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | | | - Bo Ding
- BioPharmaceuticals Medical, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
128
|
Novikova TS, Ermakov EA, Kostina EV, Sinyakov AN, Sizikov AE, Nevinsky GA, Buneva VN. Hydrolysis of Oligodeoxyribonucleotides on the Microarray Surface and in Solution by Catalytic Anti-DNA Antibodies in Systemic Lupus Erythematosus. Curr Issues Mol Biol 2023; 45:9887-9903. [PMID: 38132463 PMCID: PMC10742339 DOI: 10.3390/cimb45120617] [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: 11/02/2023] [Revised: 11/23/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Anti-DNA antibodies are known to be classical serological hallmarks of systemic lupus erythematosus (SLE). In addition to high-affinity antibodies, the autoantibody pool also contains natural catalytic anti-DNA antibodies that recognize and hydrolyze DNA. However, the specificity of such antibodies is uncertain. In addition, DNA binding to a surface such as the cell membrane, can also affect its recognition by antibodies. Here, we analyzed the hydrolysis of short oligodeoxyribonucleotides (ODNs) immobilized on the microarray surface and in solution by catalytic anti-DNA antibodies from SLE patients. It has been shown that IgG antibodies from SLE patients hydrolyze ODNs more effectively both in solution and on the surface, compared to IgG from healthy individuals. The data obtained indicate a more efficient hydrolysis of ODNs in solution than immobilized ODNs on the surface. In addition, differences in the specificity of recognition and hydrolysis of certain ODNs by anti-DNA antibodies were revealed, indicating the formation of autoantibodies to specific DNA motifs in SLE. The data obtained expand our understanding of the role of anti-DNA antibodies in SLE. Differences in the recognition and hydrolysis of surface-tethered and dissolved ODNs need to be considered in DNA microarray applications.
Collapse
Affiliation(s)
- Tatiana S. Novikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Evgeny A. Ermakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Elena V. Kostina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexander N. Sinyakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexey E. Sizikov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Sciences, 630099 Novosibirsk, Russia
| | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Valentina N. Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
129
|
Deng Y, Ou YY, Mo CJ, Huang L, Qin X. Characteristics and clustering analysis of peripheral blood lymphocyte subsets in children with systemic lupus erythematosus complicated with clinical infection. Clin Rheumatol 2023; 42:3299-3309. [PMID: 37537315 DOI: 10.1007/s10067-023-06716-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVES Clinical infection is a common complication in children with systemic lupus erythematosus (SLE). However, few studies have investigated immune alterations in children with SLE complicated with clinical infection. We assessed lymphocyte subsets in children with SLE to explore the possibility of clinical infection. METHODS We retrospectively analyzed the proportion of peripheral lymphocyte subsets in 140 children with SLE. Children with SLE were classified into different clusters according to the proportion of peripheral blood lymphocyte subsets: (CD3 + /CD4 + T cell, CD3 + /CD8 + T cell, CD3 + /CD4 + /CD8 + T cell, CD3 + /CD4-/CD8- T cell, CD19 + B cell, and CD3-/CD16 + /CD56 + NK cell). Differences in the proportion of lymphoid subsets, infection rates, and systemic lupus erythematosus disease activity index (SLEDAI) scores were compared between clusters. In addition, we grouped the subjects according to the presence or absence of infection. Proportions of lymphoid subsets, demographic variables, clinical presentation, and other laboratory variables were compared between the infected and uninfected groups. Finally, the diagnostic ability of lymphocyte subset ratios to distinguish secondary infection in children with SLE was predicted using an ROC curve. RESULTS Cluster C2 had a higher proportion of B cells than Cluster C1, while Cluster C1 had a lower proportion of NK cells, CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells. Infection rates and SLEDAI scores were higher in Cluster C2 than in Cluster C1. The infected children had a higher proportion of B cells and a lower proportion of CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells. There were no significant differences in lymphoid subsets between children in Cluster C2 and the infected groups. The area under the ROC curve of B lymphocytes in predicting SLE children with infection was 0.842. The area under the ROC curve was 0.855 when a combination of B cells, NK cells, CD4 + T cells, and CD8 + T cells was used to predict the outcome of coinfection. CONCLUSIONS A high percentage of B cells and a low percentage of CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, CD3 + /CD4 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells may be associated with infection in children with SLE. B cells was used to predict the outcome of coinfection in children with SLE. Key Points • A high percentage of B cells and a low percentage of CD3 + T cells, CD3 + /CD4 + T cells, CD3 + /CD8 + T cells, CD3 + /CD4 + /CD8 + T cells, and CD3 + /CD4-/CD8- T cells may be associated with infection in children with SLE • B cells was used to predict the outcome of coinfection in children with SLE.
Collapse
Affiliation(s)
- Yan Deng
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ying-Ying Ou
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Cui-Ju Mo
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Li Huang
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xue Qin
- Department of Clinical Laboratory, Key Laboratory of Clinical Laboratory Medicine of Guangxi, Department of Education, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
130
|
Zhi L, Gu L, Tong L, Liu X, Lu L, Guo R. Immune profile alterations of systemic lupus erythematosus patients with infections. Clin Exp Med 2023; 23:4765-4777. [PMID: 37938465 DOI: 10.1007/s10238-023-01220-z] [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: 07/13/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023]
Abstract
This study aimed to elucidate the immune status of systemic lupus erythematosus (SLE) patients with infections. We enrolled 253 SLE patients including 77 patients with infections. Clinical features and immunological parameters were analyzed, with particular reference to neutrophil CD64 (nCD64) expression, myeloid-derived suppressor cells (MDSCs), activated T cells and multiple cytokines. Among the 77 SLE patients with infections, 32 patients (41.56%) developed fever and 20 patients (25.97%) developed serositis, which were higher compared to the non-infection group. A considerably higher level of nCD64 was found in the infection group (4.65 vs 1.01, P < 0.001). In addition, the infection group exhibited higher percentages of total MDSCs (6.99 vs 4.30%, P = 0.003), polymorphonuclear MDSCs (PMN-MDSCs) (P = 0.032) and monocytic MDSCs (M-MDSCs) (P = 0.015). T cells were more activated during infections, with an elevated level of IL-2R (P < 0.001). Specifically, higher percentages of CD4+CD38+ T cells (55.73 vs 50.17%, P = 0.036), CD8+HLA-DR+ T cells (59.82 vs 47.99%, P < 0.001) and CD8+CD38+ T cells (68.59 vs 63.90%, P = 0.044) were identified in the infection group. Furthermore, the serum levels of IL-6, IL-8 and IL-10 were elevated in the infection group (all P < 0.001). Higher proportions of neutrophils, CD4+ and CD8+ T cells, and MDSCs were activated during infections in SLE patients. Additionally, the serum cytokines altered during infections, with noticeably elevated levels of IL-6, IL-8 and IL-10. Infections may lead to the amplification of immune alterations in SLE.
Collapse
Affiliation(s)
- Langxian Zhi
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200001, China
| | - Liyang Gu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200001, China
| | - Lei Tong
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiaotong University, Shanghai, 200030, China
| | - Xuesong Liu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200001, China
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Liangjing Lu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200001, China.
| | - Ruru Guo
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200001, China.
| |
Collapse
|
131
|
Sullivan KA, Chapman C, Lu L, Ashbrook DG, Wang Y, Alduraibi FK, Lu C, Sun CW, Liu S, Williams RW, Mountz JD, Hsu HC. Increased development of T-bet +CD11c + B cells predisposes to lupus in females: Analysis in BXD2 mouse and genetic crosses. Clin Immunol 2023; 257:109842. [PMID: 37981105 PMCID: PMC10799694 DOI: 10.1016/j.clim.2023.109842] [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: 07/29/2023] [Revised: 10/05/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Cardinal features of lupus include elevated B cell activation and autoantibody production with a female sex preponderance. We quantified interactions of sex and genetic variation on the development of autoimmune B-cell phenotypes and autoantibodies in the BXD2 murine model of lupus using a cohort of backcrossed progeny (BXD2 x C57BL/6J) x BXD2. Sex was the key factor leading to increased total IgG, IgG2b, and autoantibodies. The percentage of T-bet+CD11c+ IgD+ activated naive B cells (aNAV) was higher in females and was associated with increased T-bet+CD11c+ IgD- age-related B cells, Fas+GL7+ germinal center B cells, Cxcr5-Icos+ peripheral T-helper cells, and Cxcr5+Icos+ follicular T-helper cells. IFN-β was elevated in females. Variation in aNAV cells was mapped to Chr 7 in a locus that shows significant interactions between the female sex and heterozygous B/D variant. Our results suggest that activation of naive B cells forms the basis for the female-predominant development of autoantibodies in lupus-susceptible BXD2 mice.
Collapse
Affiliation(s)
- Kathryn A Sullivan
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Casey Chapman
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - David G Ashbrook
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yong Wang
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Fatima K Alduraibi
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA; Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Division of Rheumatology, Department of Medicine, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Changming Lu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chao-Wang Sun
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shanrun Liu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - John D Mountz
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA; Research, Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA
| | - Hui-Chen Hsu
- Department of Medicine, Division of Clinical Immunology and Rheumatology, the University of Alabama at Birmingham, Birmingham, AL, USA; Research, Birmingham Veterans Affairs Health Care System, Birmingham, AL, USA.
| |
Collapse
|
132
|
Waterhölter A, Wunderlich M, Turner JE. MAIT cells in immune-mediated tissue injury and repair. Eur J Immunol 2023; 53:e2350483. [PMID: 37740567 DOI: 10.1002/eji.202350483] [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: 06/23/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are T cells that express a semi-invariant αβ T-cell receptor (TCR), recognizing non-peptide antigens, such as microbial-derived vitamin B2 metabolites, presented by the nonpolymorphic MHC class I related-1 molecule. Like NKT cells and γδT cells, MAIT cells belong to the group of innate-like T cells that combine properties of the innate and adaptive immune systems. They account for up to 10% of the blood T-cell population in humans and are particularly abundant at mucosal sites. Beyond the emerging role of MAIT cells in antibacterial and antiviral defenses, increasing evidence suggests additional functions in noninfectious settings, including immune-mediated inflammatory diseases and tissue repair. Here, we discuss recent advances in the understanding of MAIT cell functions in sterile tissue inflammation, with a particular focus on autoimmunity, chronic inflammatory diseases, and tissue repair.
Collapse
Affiliation(s)
- Alex Waterhölter
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Wunderlich
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
133
|
Rojo R, Calvo Alén J, Prada Á, Valor S, Roy G, López-Hoyos M, Cervera R, Sánchez Mateos P, Jurado Roger A. Recommendations for the use of anti-dsDNA autoantibodies in the diagnosis and follow-up of systemic lupus erythematosus - A proposal from an expert panel. Autoimmun Rev 2023; 22:103479. [PMID: 37967782 DOI: 10.1016/j.autrev.2023.103479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
Anti-dsDNA autoantibodies are listed as one of the classification criteria for systemic lupus erythematosus (SLE) and are relatively effective indicators for monitoring disease activity and treatment response. Therefore, clinicians rely on them to diagnose and adjust medication and treatment strategies for SLE patients. However, the use of anti-dsDNA antibodies is not free from controversy. Part of this controversy stems from the fact that anti-dsDNA antibodies are found in several disorders, besides SLE. In addition to this, anti-dsDNA antibodies are a heterogeneous group of antibodies, and their determination still lacks proper standardization. Moreover, anti-dsDNA testing specificity and diagnostic performance change depending on the population under study. These and other issues result in inconsistency and encumber the clinical use of anti-dsDNA antibodies. A panel of medical laboratory and clinical experts on SLE discussed such issues based on their clinical experience in a first meeting, establishing a series of recommendations. The proceedings of this first meeting, plus an exhaustive review of the literature, were used to compose a paper draft. The panel subsequently discussed and refined this draft in a second meeting, the result of which is this paper. This document is relevant to clinical laboratories as it guides to improving diagnosis and monitoring of SLE. Simultaneously, it will help laboratories compile more informative reports, not limited to a mere number. It is also relevant to clinical doctors who wish to better understand laboratory methods so that they can do a more efficient, better-aimed laboratory test ordering.
Collapse
Affiliation(s)
- Ricardo Rojo
- Specialist Consultant at the Immunology Department of the University Hospital of A Coruña, Spain
| | - Jaime Calvo Alén
- Head of the Rheumatology Department at the Araba University Hospital, Vitoria, Spain
| | - Álvaro Prada
- Head of Section at the Immunology Laboratory of the University Hospital of Donostia, Spain
| | | | - Garbiñe Roy
- Head of the Autoimmunity Section at the Immunology Department of the Ramón y Cajal University Hospital, Madrid, Spain
| | - Marcos López-Hoyos
- Head of the Immunology Department at the Marqués de Valdecilla-IDIVAL University Hospital, Santander. Full Professor, Molecular Biology Department at the University of Cantabria, Santander, Spain.
| | - Ricard Cervera
- Head of the Department of Autoimmune Diseases, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Paloma Sánchez Mateos
- Full Professor at the Complutense University, and Specialist Consultant at the Immunology Department of the Gregorio Marañón General University Hospital, Madrid, Spain
| | - Aurora Jurado Roger
- Head of Section at the Immunology and Allergology Department of the Reina Sofía University Hospital, Córdoba, Spain
| |
Collapse
|
134
|
Falkenstein DK, Jarvis JN. Systemic lupus erythematosus in American Indian/Alaska natives: Incorporating our new understanding of the biology of trauma. Semin Arthritis Rheum 2023; 63:152245. [PMID: 37595507 DOI: 10.1016/j.semarthrit.2023.152245] [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: 01/11/2023] [Revised: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 08/20/2023]
Abstract
OBJECTIVE To review the literature regarding systemic lupus erythematosus (SLE) in American Indian/Alaska Native (AI/AN) people and relate prevalence and/or disease severity to our emerging understanding of the biology of trauma and toxic stress. METHODS We conducted a search and review of the literature using search terms "lupus and American Indians" "ACEs and disease outcome" "Biology of Adversity" "lupus and ACE scores," " lupus and childhood abuse." These search criteria were entered into Google Scholar and articles retrieved from PubMed, NBCI. This approach yielded a small numbers of papers used throughout this review. We excluded articles that were not published in a peer reviewed journals, as well as editorial commentaries. RESULTS In the AI/AN population, SLE shows high prevalence rates and severe disease manifestations, comparable to the African American population. AI/AN populations also have high rates of childhood trauma. Toxic stress and trauma such as those catalogued in the Adverse Childhood Experiences (ACE) study have broad-reaching immunologic and epigenetic effects that are likely to be relevant to our understanding of SLE in AI/AN people. CONCLUSIONS AI/AN people have high rates of SLE. These high rates are likely to be driven by many complex factors, not all of which are genetic. Future research is needed to establish (or refute) a causal connection between the biology of adversity and SLE in socially marginalized and historically traumatized populations.
Collapse
Affiliation(s)
- Danielle K Falkenstein
- Medical Student, University at Buffalo Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY, USA
| | - James N Jarvis
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY, USA; Genetics, Genomics, & Bioinformatics Program, University at Buffalo Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY, USA.
| |
Collapse
|
135
|
Kalayci FNC, Ozen S. Possible Role of Dysbiosis of the Gut Microbiome in SLE. Curr Rheumatol Rep 2023; 25:247-258. [PMID: 37737528 DOI: 10.1007/s11926-023-01115-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 09/23/2023]
Abstract
PURPOSE OF REVIEW The resident gut microbiota serves as a double-edged sword that aids the host in multiple ways to preserve a healthy equilibrium and serve as early companions and boosters for the gradual evolution of our immune defensive layers; nevertheless, the perturbation of the symbiotic resident intestinal communities has a profound impact on autoimmunity induction, particularly in systemic lupus erythematosus (SLE). Herein, we seek to critically evaluate the microbiome research in SLE with a focus on intestinal dysbiosis. RECENT FINDINGS SLE is a complex and heterogeneous disorder with self-attack due to loss of tolerance, and there is aberrant excessive immune system activation. There is mounting evidence suggesting that intestinal flora disturbances may accelerate the formation and progression of SLE, presumably through a variety of mechanisms, including intestinal barrier dysfunction and leaky gut, molecular mimicry, bystander activation, epitope spreading, gender bias, and biofilms. Gut microbiome plays a critical role in SLE pathogenesis, and additional studies are warranted to properly define the impact of gut microbiome in SLE, which can eventually lead to new and potentially safer management approaches for this debilitating disease.
Collapse
Affiliation(s)
| | - Seza Ozen
- Department of Paediatric Rheumatology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
136
|
Carlsson E, Cowell-McGlory T, Hedrich CM. cAMP responsive element modulator α promotes effector T cells in systemic autoimmune diseases. Immunology 2023; 170:470-482. [PMID: 37435993 DOI: 10.1111/imm.13680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/01/2023] [Indexed: 07/13/2023] Open
Abstract
T lymphocytes play a crucial role in adaptive immunity. Dysregulation of T cell-derived inflammatory cytokine expression and loss of self-tolerance promote inflammation and tissue damage in several autoimmune/inflammatory diseases, including systemic lupus erythematosus (SLE) and psoriasis. The transcription factor cAMP responsive element modulator α (CREMα) plays a key role in the regulation of T cell homeostasis. Increased expression of CREMα is a hallmark of the T cell-mediated inflammatory diseases SLE and psoriasis. Notably, CREMα regulates the expression of effector molecules through trans-regulation and/or the co-recruitment of epigenetic modifiers, including DNA methyltransferases (DNMT3a), histone-methyltransferases (G9a) and histone acetyltransferases (p300). Thus, CREMα may be used as a biomarker for disease activity and/or target for future targeted therapeutic interventions.
Collapse
Affiliation(s)
- Emil Carlsson
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Taylor Cowell-McGlory
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
- Paediatric Excellence Initiative, NIHR Great Ormond Street Biomedical Research Centre, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| |
Collapse
|
137
|
Gracia-Ramos AE, Saavedra MA. Systemic lupus erythematosus after SARS-CoV-2 infection: A causal or temporal relationship? Int J Rheum Dis 2023; 26:2373-2376. [PMID: 38041650 DOI: 10.1111/1756-185x.14896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 12/03/2023]
Affiliation(s)
- Abraham Edgar Gracia-Ramos
- Departamento de Medicina Interna, Hospital General, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Miguel Angel Saavedra
- División de Investigación en Salud, Hospital de Especialidades Dr. Antonio Fraga Mouret, Centro Médico Nacional La Raza, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
138
|
Hua N, Gomez A, Lindblom J, Emamikia S, Enman Y, Grannas D, Heintz E, Regardt M, Parodis I. Sensitivity analysis of EQ-5D-3L index scores in terms of discriminative and known-groups validity in SLE: introducing Adequate Health State. Rheumatology (Oxford) 2023; 62:3916-3923. [PMID: 36946293 PMCID: PMC10691928 DOI: 10.1093/rheumatology/kead140] [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: 01/24/2023] [Revised: 02/28/2023] [Accepted: 03/18/2023] [Indexed: 03/23/2023] Open
Abstract
OBJECTIVES To investigate the ability of different EuroQol 5-Dimensions 3-Levels (EQ-5D-3L) index scores to discriminate between verum drug and placebo (discriminant validity) as well as between responders and non-responders (known-groups validity) in the SLE patient population of two phase III clinical trials of belimumab. METHODS Data from the BLISS-52 (NCT00424476) and BLISS-76 (NCT00410384) trials (N = 1684), which both showed superiority of belimumab to placebo, were utilized. Responders were defined as SLE Responder Index 4 (SRI-4) achievers at week 52. The Pearson's χ2 and Mann-Whitney U tests were used for comparisons, and logistic regression analysis was used for adjustments for confounders and assessment of independence. RESULTS While full health state (FHS; EQ-5D index score 1) showed the best ability to discriminate between belimumab and placebo [adjusted odds ratio (OR) 1.47; 95% CI 1.11, 1.96; P = 0.008] and between SRI-4 responders and non-responders (adjusted OR 3.47; 95% CI 1.29, 10.98; P = 0.020), the discriminative ability of EQ-5D index scores 0.800 or more reached statistical significance for both discriminant validity (adjusted OR 1.29; 95% CI 1.02, 1.63; P = 0.036) and known-groups validity (adjusted OR 3.08; 95% CI 1.16, 9.69; P = 0.034). CONCLUSION Overall, higher EQ-5D index scores were associated with increasing ability to discriminate between belimumab and placebo, and between responders and non-responders. EQ-5D index scores less stringent than FHS may be clinically relevant health-related quality of life goals of treatment in patients with SLE, introducing the concept of EQ-5D adequate health state when FHS is not achievable.
Collapse
Affiliation(s)
- Nicole Hua
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Alvaro Gomez
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Sharzad Emamikia
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Yvonne Enman
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- The Swedish Rheumatism Association, Stockholm, Sweden
| | - David Grannas
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emelie Heintz
- Department of Learning, Informatics, Management and Ethics (LIME), Karolinska Institutet, Stockholm, Sweden
| | - Malin Regardt
- Department of Neurobiology, Care Sciences and Society, Division of Occupational Therapy, Karolinska Institutet, Stockholm, Sweden
- Women’s Health and Allied Health Professionals Theme, Medical Unit Occupational Therapy and Physiotherapy, Karolinska University Hospital, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
139
|
Eryavuz Onmaz D, Tezcan D, Yilmaz S, Onmaz M, Unlu A. Altered kynurenine pathway metabolism and association with disease activity in patients with systemic lupus. Amino Acids 2023; 55:1937-1947. [PMID: 37925676 DOI: 10.1007/s00726-023-03353-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease accompanied by increased release of proinflammatory cytokines that are known to activate the indoleamine 2,3-dioxygenase (IDO-1) enzyme, which catalyzes the rate-limiting step of the kynurenine pathway (KP). This study aimed to measure KP metabolite levels in patients with SLE and investigate the relationship between disease activity, clinical findings, and KP. The study included 100 patients with SLE and 100 healthy controls. Serum tryptophan (TRP), kynurenine (KYN), kynurenic acid (KYNA), 3-hydroxyanthranilic acid (3HAA), 3-hydroxykynurenine (3HK), quinolinic acid (QA) concentrations were measured with tandem mass spectrometry. Serum KYN, KYNA, 3HAA, 3HK, and QA levels of the patients with SLE were significantly higher than the control group. Serum QA levels were elevated in patients with neurological involvement (four patients with peripheral neuropathy and two patients with mononeuropathy), serum KYN levels and KYN/TRP ratio increased in patients with joint involvement, and serum KYN, 3HK, and 3HAA levels and the KYN/TRP ratio were increased in patients with renal involvement. Moreover, KYN and KYN/TRP ratios were positively correlated with the disease activity score. These findings indicated that imbalances in KP metabolites may be associated with the pathogenesis, activation, and clinical manifestations of SLE.
Collapse
Affiliation(s)
- Duygu Eryavuz Onmaz
- Department of Biochemistry, Selcuk University Faculty of Medicine, Alaaddin Keykubat Campus, Selcuklu, 42075, Konya, Turkey.
| | - Dilek Tezcan
- Division of Rheumatology, Gülhane Training and Research Hospital, Ankara, Turkey
| | - Sema Yilmaz
- Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Mustafa Onmaz
- Faculty of Medicine, Department of Family Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Ali Unlu
- Department of Biochemistry, Selcuk University Faculty of Medicine, Alaaddin Keykubat Campus, Selcuklu, 42075, Konya, Turkey
| |
Collapse
|
140
|
Bekaryssova D, Mruthyunjaya Vijaya P, Ahmed S, Sondur S, Zimba O. Revisiting articular syndrome in the peri-pandemic COVID-19 era. Rheumatol Int 2023; 43:2157-2166. [PMID: 37747562 DOI: 10.1007/s00296-023-05459-3] [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: 08/23/2023] [Accepted: 09/03/2023] [Indexed: 09/26/2023]
Abstract
Articular syndrome is often the presentation of a person's various rheumatic or related diseases. It includes both arthralgia and arthritis, with objective signs of joint inflammation defining the latter. This syndromic approach to joint pain enables a scientific method for early diagnosis of common rheumatic conditions without compromising the recognition of uncommon conditions. This review explores common rheumatic conditions associated with articular syndrome, including osteoarthritis, rheumatoid arthritis (RA), systemic lupus erythematosus (SLE). It supports the early differentiation of uncommon but emerging entities such as reactive arthritis (ReA). The aim of the review is to comprehensively overview various forms of articular syndrome to update rheumatologists' and allied health specialists' knowledge. Epidemiology, clinical presentations, diagnostic approaches, and therapeutic strategies are discussed in the context of articular syndrome. The challenges emerging in the peri-pandemic COVID-19 era are highlighted. The improved understanding of the spectrum of clinical conditions and disease states presenting with articular syndrome may facilitate early diagnosis, optimal management, and enhanced patient outcomes within the realm of rheumatology.
Collapse
Affiliation(s)
- Dana Bekaryssova
- Department of Biology and Biochemistry, South Kazakhstan Medical Academy, Shymkent, Kazakhstan.
| | - Prakashini Mruthyunjaya Vijaya
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, India
| | - Sakir Ahmed
- Department of Clinical Immunology and Rheumatology, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, India
| | - Suhas Sondur
- Department of Orthopedics, Kalinga Institute of Medical Sciences, KIIT University, Bhubaneswar, India
| | - Olena Zimba
- Department of Clinical Rheumatology and Immunology, University Hospital in Krakow, Kraków, Poland
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Department of Internal Medicine N2, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
141
|
Zhang L, Du F, Jin Q, Sun L, Wang B, Tan Z, Meng X, Huang B, Zhan Y, Su W, Song R, Wu C, Chen L, Chen X, Ding X. Identification and Characterization of CD8 + CD27 + CXCR3 - T Cell Dysregulation and Progression-Associated Biomarkers in Systemic Lupus Erythematosus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300123. [PMID: 37875396 PMCID: PMC10724430 DOI: 10.1002/advs.202300123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/04/2023] [Indexed: 10/26/2023]
Abstract
Systemic Lupus Erythematosus (SLE) etiopathogenesis highlights the contributions of overproduction of CD4+ T cells and loss of immune tolerance. However, the involvement of CD8+ T cells in SLE pathology and disease progression remains unclear. Here, the comprehensive immune cell dysregulation in total 263 clinical peripheral blood samples composed of active SLE (aSLE), remission SLE (rSLE) and healthy controls (HCs) is investigated via mass cytometry, flow cytometry and single-cell RNA sequencing. This is observed that CD8+ CD27+ CXCR3- T cells are increased in rSLE compare to aSLE. Meanwhile, the effector function of CD8+ CD27+ CXCR3- T cells are overactive in aSLE compare to HCs and rSLE, and are positively associated with clinical SLE activity. In addition, the response of peripheral blood mononuclear cells (PBMCs) is monitored to interleukin-2 stimulation in aSLE and rSLE to construct dynamic network biomarker (DNB) model. It is demonstrated that DNB score-related parameters can faithfully predict the remission of aSLE and the flares of rSLE. The abundance and functional dysregulation of CD8+ CD27+ CXCR3- T cells can be potential biomarkers for SLE prognosis and concomitant diagnosis. The DNB score with accurate prediction to SLE disease progression can provide clinical treatment suggestions especially for drug dosage determination.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200001China
| | - Fang Du
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200001China
| | - Qiqi Jin
- Key Laboratory of Systems BiologyCenter for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- University of Chinese Academy of SciencesBeijing100049China
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
| | - Li Sun
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhou325000China
| | - Boqian Wang
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200001China
| | - Ziyang Tan
- Science for Life LaboratoryDepartment of Women's and Children's HealthKarolinska InstitutetSolna17121Sweden
| | - Xinyu Meng
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200001China
| | - Baozhen Huang
- Department of Chemical PathologyLi Ka Shing Institute of Health SciencesFaculty of MedicineThe Chinese University of Hong KongHong Kong999077China
| | - Yifan Zhan
- Drug DiscoveryShanghai Huaota Biopharmaceutical Co. Ltd.Shanghai200131China
| | - Wenqiong Su
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200001China
| | - Rui Song
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
- Nantong First People's HospitalAffiliated Hospital 2 of Nantong UniversityNantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine
| | - Chunmei Wu
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
| | - Luonan Chen
- Key Laboratory of Systems BiologyCenter for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesShanghai200031China
- School of Life Science and TechnologyShanghaiTech UniversityShanghai201210China
- Key Laboratory of Systems Health Science of Zhejiang ProvinceSchool of Life ScienceHangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesChinese Academy of SciencesHangzhou310024China
| | - Xiaoxiang Chen
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
| | - Xianting Ding
- Department of RheumatologyShanghai Jiao Tong University School of Medicine Affiliated Renji Hospital and School of Biomedical EngineeringShanghai200030China
- State Key Laboratory of Oncogenes and Related GenesInstitute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200001China
| |
Collapse
|
142
|
Liu F, Xie Y, Wu C, Liu H, Zhang J, Lian Y, Xie N. A nested case-control study investigating short-term prognosis, clinical and imaging features in acute ischemic stroke patients with systemic lupus erythematosus. Acta Neurol Belg 2023; 123:2177-2184. [PMID: 36719615 DOI: 10.1007/s13760-023-02189-y] [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: 04/10/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVES To investigate acute ischemic stroke (AIS) patients with systemic lupus erythematosus (SLE) clinical and imaging features, and to explore the impact of SLE on the short-term prognosis of AIS patients. METHODS A nested case-control study was conducted in The First Affiliated Hospital of Zhengzhou University between October 1, 2019, and May 31, 2021. The case group consisted of 28 AIS patients diagnosed with SLE, and 112 AIS patients without SLE were selected by incidence density sampling as a control group. RESULTS Among 140 patients, the mean age was (48.4 ± 15.6) years, and 130 (92.9%) were females. Higher levels in low-density lipoprotein cholesterol (LDL-C) (2.5 mmol/L vs. 2.1 mmol/L; P = 0.049), D-dimer (DDi) (0.7 mg/L vs. 0.3 mg/L; P = 0.02), and C-reactive protein (CRP) (13.2 mg/L vs. 6.3 mg/L; P = 0.002) were observed in the case group. On imaging, the case group was more prevalent in simultaneous involvement of the anterior and posterior circulation (42.9% vs. 17.0%; P = 0.004), multiple infarcts (46.4% vs. 20.5%; P = 0.008) and stroke of other undetermined etiologies (SUE) (28.6% vs. 6.3%; P = 0.002) than the control group. SLE (OR 5.94, 95%CI [1.04-34.39]; P = 0.045) was an independent risk factor for a poor short-term prognosis of AIS patients. CONCLUSIONS Higher levels of LDL-C, CRP, and DDi, multiple infarcts and simultaneous involvement of the anterior, and posterior circulation were more prevalent in the AIS patients with SLE. Further, SLE was also found as an independent risk factor for AIS patients' poor short-term prognosis.
Collapse
Affiliation(s)
- Fengxia Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Yinyin Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Chuanjie Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Hongbo Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Jiwei Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Nanchang Xie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
143
|
Lindblom J, Beretta L, Borghi MO, Alarcón-Riquelme ME, Parodis I. Serum profiling identifies CCL8, CXCL13, and IL-1RA as markers of active disease in patients with systemic lupus erythematosus. Front Immunol 2023; 14:1257085. [PMID: 38098483 PMCID: PMC10720584 DOI: 10.3389/fimmu.2023.1257085] [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: 07/11/2023] [Accepted: 11/13/2023] [Indexed: 12/17/2023] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is a clinically heterogeneous disease that presents a challenge for clinicians. To identify potential biomarkers for diagnosis and disease activity in SLE, we investigated a selected yet broad panel of cytokines and autoantibodies in patients with SLE, healthy controls (HC), and patients with other autoimmune diseases (AIDs). Methods Serum samples from 422 SLE patients, 546 HC, and 1223 other AIDs were analysed within the frame of the European PRECISESADS project (NTC02890121). Cytokine levels were determined using Luminex panels, and autoantibodies using different immunoassays. Results Of the 83 cytokines analysed, 29 differed significantly between patients with SLE and HC. Specifically, CCL8, CXCL13, and IL-1RA levels were elevated in patients with active, but not inactive, SLE versus HC, as well as in patients with SLE versus other AIDs. The levels of these cytokines also correlated with SLE Disease Activity Index 2000 (SLEDAI-2K) scores, among five other cytokines. Overall, the occurrence of autoantibodies was similar across SLEDAI-2K organ domains, and the correlations between autoantibodies and activity in different organ domains were weak. Discussion Our findings suggest that, upon validation, CCL8, CXCL13, and IL-1RA could serve as promising serum biomarkers of activity in SLE.
Collapse
Affiliation(s)
- Julius Lindblom
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Beretta
- Referral Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Maria Orietta Borghi
- IRCCS Istituto Auxologico Italiano, Immunorheumatology Research Laboratory, Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Marta E. Alarcón-Riquelme
- GENYO, Centre for Genomics and Oncological Research: Pfizer, University of Granada/Andalusian Regional Government, Medical Genomics, Granada, Spain
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis Parodis
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden
- Department of Rheumatology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
144
|
Kubota T. An Emerging Role for Anti-DNA Antibodies in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:16499. [PMID: 38003689 PMCID: PMC10671047 DOI: 10.3390/ijms242216499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Anti-DNA antibodies are hallmark autoantibodies produced in systemic lupus erythematosus (SLE), but their pathogenetic role is not fully understood. Accumulating evidence suggests that some anti-DNA antibodies enter different types of live cells and affect the pathophysiology of SLE by stimulating or impairing these cells. Circulating neutrophils in SLE are activated by a type I interferon or other stimuli and are primed to release neutrophil extracellular traps (NETs) on additional stimulation. Anti-DNA antibodies are also involved in this process and may induce NET release. Thereafter, they bind and protect extracellular DNA in the NETs from digestion by nucleases, resulting in increased NET immunogenicity. This review discusses the pathogenetic role of anti-DNA antibodies in SLE, mainly focusing on recent progress in the two research fields concerning antibody penetration into live cells and NETosis.
Collapse
Affiliation(s)
- Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Tsuchiura 300-0051, Ibaraki, Japan
| |
Collapse
|
145
|
Zhu T, Ding Y, Xu X, Zhang L, Zhang X, Cui Y, Liu L. Trans-ethnic Mendelian randomization study of systemic lupus erythematosus and common female hormone-dependent malignancies. Chin Med J (Engl) 2023; 136:2609-2620. [PMID: 37027287 PMCID: PMC10617913 DOI: 10.1097/cm9.0000000000002555] [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: 12/06/2022] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Observational research has reported that systemic lupus erythematosus (SLE) is related to common female hormone-dependent cancers, but the underlying causal effect remains undefined. This study aimed to explore the causal association of these conditions by Mendelian randomization (MR) analysis. METHODS We selected instrumental variables for SLE from genome-wide association studies (GWASs) conducted in European and East Asian populations. The genetic variants for female malignant neoplasms were obtained from corresponding ancestry GWASs. We utilized inverse variance weighted (IVW) as the primary analysis, followed by sensitivity analysis. Furthermore, we conducted multivariable MR (MVMR) to estimate direct effects by adjusting for the body mass index and estradiol. Finally, we implemented reverse direction MR analysis and gave a negative example to test the reliability of MR results. RESULTS We found SLE was significantly negatively associated with overall endometrial cancer risk (odds ratio [OR] = 0.961, 95% confidence interval [CI] = 0.935-0.987, P = 3.57E-03) and moderately inversely related to endometrioid endometrial cancer (ENEC) (OR = 0.965, 95% CI = 0.936-0.995, P = 0.024) risk in the European population by IVW. We replicated these results using other MR models and detected a direct effect by MVMR (overall endometrial cancer, OR = 0.962, 95% CI = 0.941-0.983, P = 5.11E-04; ENEC, OR = 0.964, 95% CI = 0.940-0.989, P = 0.005). Moreover, we revealed that SLE was correlated with decreased breast cancer risk (OR = 0.951, 95% CI = 0.918-0.986, P = 0.006) in the East Asian population by IVW, and the effect was still significant in MVMR (OR = 0.934, 95% CI = 0.859-0.976, P = 0.002). The statistical powers of positive MR results were all >0.9. CONCLUSION This finding suggests a possible causal effect of SLE on the risk of overall endometrial cancer and breast cancer in European and East Asian populations, respectively, by MR analysis, which compensates for inherent limitations of observational research.
Collapse
Affiliation(s)
- Tingting Zhu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
- Department of Rheumatology and Immunology, Arthritis Research Institute, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Yantao Ding
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Xiaoli Xu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Liyin Zhang
- Department of Dermatology, Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214001, China
| | - Xuejun Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| | - Yong Cui
- Department of Dermatology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Lu Liu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- Institute of Dermatology, Anhui Medical University, Hefei, Anhui 230022, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui 230022, China
| |
Collapse
|
146
|
Tan B, So PN, Krishnan A, Carriazo S, Bahamonde JR, Lamech TM, Hassanein M, Lerma E, Wiegley N. Approach to Pregnancy in Patients With Lupus Nephritis. Kidney Med 2023; 5:100724. [PMID: 37915962 PMCID: PMC10616386 DOI: 10.1016/j.xkme.2023.100724] [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] [Indexed: 11/03/2023] Open
Abstract
Active lupus nephritis (LN) in pregnancy is strongly associated with poor maternal and fetal outcomes and, therefore, has implications on the planning, timing, and management. Prepregnancy evaluation is essential for all LN patients with childbearing potential to ensure pregnancies proceed in a safe and timely manner. Both maternal and fetal risks are communicated to patient during the evaluation. Stratification into different risk profile groups is then made based on disease activity and organ impairment severity. Patients with LN are generally divided into 3 main groups. Patients with LN who become pregnant receive treatments that are nonteratogenic and optimal for fetal and maternal outcomes. Throughout the pregnancy period, these patients are monitored closely under surveillance by a multidisciplinary team of clinicians. The management of patients with LN in pregnancy can be challenging both diagnostically (distinguishing LN from pre-eclampsia and determining the role and timing of kidney biopsy) and therapeutically (LN flares during pregnancy and managing a newly diagnosed LN during pregnancy).
Collapse
Affiliation(s)
- Benjamin Tan
- Nephrology Unit, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia
| | | | | | - Sol Carriazo
- Department of Nephrology and Hypertension Department, Fundacion Jimenez Diaz Hospital, Madrid, Spain
| | | | | | - Mohamed Hassanein
- Division of Nephrology and Hypertension, University of Mississippi Medical Center, Jackson, MI
| | - Edgar Lerma
- Section of Nephrology, University of Illinois at Chicago, Chicago, IL
| | - Nasim Wiegley
- Division of Nephrology, University of California Davis School of Medicine, Sacramento, CA
| | - GlomCon Editorial Team
- Nephrology Unit, Queen Elizabeth Hospital, Kota Kinabalu, Malaysia
- Private Practice, Manila, Philippines
- Royal Perth Hospital, Perth, Australia
- Department of Nephrology and Hypertension Department, Fundacion Jimenez Diaz Hospital, Madrid, Spain
- Department of Nephrology, St Luke’s University Health Network, PA
- Institute of Nephrology, Madras Medical College, India
- Division of Nephrology and Hypertension, University of Mississippi Medical Center, Jackson, MI
- Section of Nephrology, University of Illinois at Chicago, Chicago, IL
- Division of Nephrology, University of California Davis School of Medicine, Sacramento, CA
| |
Collapse
|
147
|
Liu C, Wang Y, Zhang YH, Yuan Z, Zhang Z, Zeng X, Guan Z, Bahabayi A, Lu S. Elevated Layilin-Positive Monocyte Levels in the Peripheral Blood of Patients with Systemic Lupus Erythematosus Reflect Their Autoimmune Status. Immunol Invest 2023; 52:879-896. [PMID: 37642473 DOI: 10.1080/08820139.2023.2249531] [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: 08/31/2023]
Abstract
OBJECTIVE To investigate the expression of layilin (LAYN) in human circulating monocytes and lymphocytes and its clinical significance in systemic lupus erythematosus (SLE). METHODS Blood samples were collected from 51 SLE patients and 50 healthy controls. Flow cytometry was used to analyze LAYN in lymphocytes and monocyte subsets. Functionally characterized molecules including human HLA, CD74 and CD62L were studied in LAYN+ monocytes. A correlation analysis was conducted between LAYN-related subsets and clinical indicators of SLE such as anti-double-stranded DNA and complements levels. ROC curves were used to explore the potential clinical diagnostic value of LAYN in SLE. RESULTS LAYN was significantly higher in monocytes than in lymphocytes and higher in CD14+CD16+ monocytes than in CD14-CD16+ and CD14+CD16- monocytes. CD74 was upregulated and CD62L was downregulated in LAYN+ monocytes compared with LAYN- monocytes. The absolute number of LAYN+ monocytes was increased in SLE patients, and the median fluorescence intensity of HLA was decreased. LAYN+ monocytes were positively correlated with complement C4, while decreased CD62L+ percentages in LAYN+ monocytes were negatively correlated with C4. The ROC analysis revealed that the area under the curve (AUCs) for CD62L+ percentages in LAYN+ monocytes, LAYN+ lymphocyte numbers, and LAYN+ monocyte numbers to distinguish SLE from healthy individuals were 0.6245, 0.6196 and 0.6173, respectively. CONCLUSION LAYN is differentially expressed in monocytes and their subpopulations and has corresponding functional differences. Changes in LAYN expression on monocytes are associated with complement C4 levels in SLE patients. These suggest that LAYN may be involved in the pathogenesis of SLE. ABBREVIATION ANOVA: analysis of variance; anti-dsDNA: anti-double-stranded DNA; anti-ENA: anti-extractable nuclear antigen; anti-SSA: anti-Sjogren syndrome A; anti-SSB: anti-Sjogren syndrome B; anti-U1RNP: anti-U1 ribonucleoprotein; AUC: area under the ROC curve; CBC: complete blood count; CD62L: L-selectin; CD74/Ii: MHC class II invariant chain; CD44/HCAM: homing cell adhesion molecule; cMos: classical monocytes; CRP: C-reactive protein; CXCR2: C-X-C motif chemokine receptor 2; CXCR4: C-X-C motif chemokine receptor 4; ESR: erythrocyte sedimentation rate; HCs: healthy controls; HA: hyaluronan; HLA: human leukocyte antigen; Ig: immunoglobulin; iMos: intermediate monocytes; LAYN: layilin; MFI: median fluorescence intensity; MIF: migration inhibitory factor; ncMos: nonclassical monocytes; PBMCs: peripheral blood mononuclear cells; ROC: receiver operating characteristic curve; SLE: systemic lupus erythematosus; SLEDAI, SLE disease activity index; Treg: regulatory T cells; WBCs: white blood cells.
Collapse
Affiliation(s)
- Chen Liu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Yiying Wang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ya-Hui Zhang
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zihang Yuan
- 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
| | - Xingyue Zeng
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Zhao Guan
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Ayibaota Bahabayi
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| | - Songsong Lu
- Department of Clinical Laboratory, Peking University People's Hospital, Beijing, China
| |
Collapse
|
148
|
Allam AR, Alhateem MS, Mahmoud AM. Efficacy and safety of baricitinib in treatment of systemic lupus erythematosus: a systematic review and meta-analysis. BMC Rheumatol 2023; 7:40. [PMID: 37904221 PMCID: PMC10617176 DOI: 10.1186/s41927-023-00363-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/10/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND SLE is an autoimmune disease marked by broad immunological dysregulation and multi-system inflammation. Baricitinib is one of the novel treatments for SLE. We conducted this meta-analysis to evaluate its safety and effectiveness in treating SLE. METHOD We looked for all published randomized controlled trials in PubMed, Scopus, Web of Science, and Cochrane and included all RCTs comparing baricitinib and placebo in the treatment of SLE. Review Manager 5.4 program was used for data analysis. RESULTS Three trials with a total of 1849 individuals were included. Participants in the baricitinib group were significantly more likely to attain SRI-4 response than those in the placebo group [RR = 1.11, 95% CI (1.02, 1.21), P = 0.01]. Additionally, baricitinib performed better than the placebo in terms of reduction of ≥ 4 points from baseline in SLEDAI-2 K score [RR = 1.13, 95% CI (1.04, 1.22), P = 0.004]. In terms of SLEDAI-2 K remission of arthritis or rash, baricitinib was also superior to placebo [RR = 1.08, 95% CI (1.00, 1.17), P = 0.04]. Treatment-emergent adverse events did not differ significantly [RR = 1.01, 95% CI (0.97, 1.05), P = 0.61]. CONCLUSION Baricitinib is potentially safe and effective in the treatment of SLE. It has successfully met the study's primary endpoint and some secondary endpoints highlighting its potential to improve the outcomes of SLE. Despite achieving an SRI-4 response, glucocorticoids sparing and some other secondary outcomes weren't reached by baricitinib.
Collapse
Affiliation(s)
- Abdallah R Allam
- Faculty of Medicine, Menoufia University, Yassin Abdelghaffar Street from Gamal Abdelnaser Street. Shebin Al- Kom, Menoufia, 32511, Egypt.
| | - Mohamed Salah Alhateem
- Faculty of Medicine, Menoufia University, Yassin Abdelghaffar Street from Gamal Abdelnaser Street. Shebin Al- Kom, Menoufia, 32511, Egypt
| | - Abdelrahman Mohamed Mahmoud
- Faculty of Medicine, Menoufia University, Yassin Abdelghaffar Street from Gamal Abdelnaser Street. Shebin Al- Kom, Menoufia, 32511, Egypt
| |
Collapse
|
149
|
Shoda H, Natsumoto B, Fujio K. Investigation of immune-related diseases using patient-derived induced pluripotent stem cells. Inflamm Regen 2023; 43:51. [PMID: 37876023 PMCID: PMC10594759 DOI: 10.1186/s41232-023-00303-4] [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: 08/08/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023] Open
Abstract
The precise pathogenesis of immune-related diseases remains unclear, and new effective therapeutic choices are required for the induction of remission or cure in these diseases. Basic research utilizing immune-related disease patient-derived induced pluripotent stem (iPS) cells is expected to be a promising platform for elucidating the pathogenesis of the diseases and for drug discovery. Since autoinflammatory diseases are usually monogenic, genetic mutations affect the cell function and patient-derived iPS cells tend to exhibit disease-specific phenotypes. In particular, iPS cell-derived monocytic cells and macrophages can be used for functional experiments, such as inflammatory cytokine production, and are often employed in research on patients with autoinflammatory diseases.On the other hand, the utilization of disease-specific iPS cells is less successful for research on autoimmune diseases. One reason for this is that autoimmune diseases are usually polygenic, which makes it challenging to determine which factors cause the phenotypes of patient-derived iPS cells are caused by. Another reason is that protocols for differentiating some lymphocytes associated with autoimmunity, such as CD4+T cells or B cells, from iPS cells have not been well established. Nevertheless, several groups have reported studies utilizing autoimmune disease patient-derived iPS cells, including patients with rheumatoid arthritis, systemic lupus erythematosus (SLE), and systemic sclerosis. Particularly, non-hematopoietic cells, such as fibroblasts and cardiomyocytes, differentiated from autoimmune patient-derived iPS cells have shown promising results for further research into the pathogenesis. Recently, our groups established a method for differentiating dendritic cells that produce interferon-alpha, which can be applied as an SLE pathological model. In summary, patient-derived iPS cells can provide a promising platform for pathological research and new drug discovery in the field of immune-related diseases.
Collapse
Affiliation(s)
- Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| | - Bunki Natsumoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehirocho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| |
Collapse
|
150
|
Tahara S, Tsuchiya T, Matsumoto H, Ozaki H. Transcription factor-binding k-mer analysis clarifies the cell type dependency of binding specificities and cis-regulatory SNPs in humans. BMC Genomics 2023; 24:597. [PMID: 37805453 PMCID: PMC10560430 DOI: 10.1186/s12864-023-09692-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/21/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND Transcription factors (TFs) exhibit heterogeneous DNA-binding specificities in individual cells and whole organisms under natural conditions, and de novo motif discovery usually provides multiple motifs, even from a single chromatin immunoprecipitation-sequencing (ChIP-seq) sample. Despite the accumulation of ChIP-seq data and ChIP-seq-derived motifs, the diversity of DNA-binding specificities across different TFs and cell types remains largely unexplored. RESULTS Here, we applied MOCCS2, our k-mer-based motif discovery method, to a collection of human TF ChIP-seq samples across diverse TFs and cell types, and systematically computed profiles of TF-binding specificity scores for all k-mers. After quality control, we compiled a set of TF-binding specificity score profiles for 2,976 high-quality ChIP-seq samples, comprising 473 TFs and 398 cell types. Using these high-quality samples, we confirmed that the k-mer-based TF-binding specificity profiles reflected TF- or TF-family dependent DNA-binding specificities. We then compared the binding specificity scores of ChIP-seq samples with the same TFs but with different cell type classes and found that half of the analyzed TFs exhibited differences in DNA-binding specificities across cell type classes. Additionally, we devised a method to detect differentially bound k-mers between two ChIP-seq samples and detected k-mers exhibiting statistically significant differences in binding specificity scores. Moreover, we demonstrated that differences in the binding specificity scores between k-mers on the reference and alternative alleles could be used to predict the effect of variants on TF binding, as validated by in vitro and in vivo assay datasets. Finally, we demonstrated that binding specificity score differences can be used to interpret disease-associated non-coding single-nucleotide polymorphisms (SNPs) as TF-affecting SNPs and provide candidates responsible for TFs and cell types. CONCLUSIONS Our study provides a basis for investigating the regulation of gene expression in a TF-, TF family-, or cell-type-dependent manner. Furthermore, our differential analysis of binding-specificity scores highlights noncoding disease-associated variants in humans.
Collapse
Affiliation(s)
- Saeko Tahara
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
- School of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Takaho Tsuchiya
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
- Center for Artificial Intelligence Research, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Hirotaka Matsumoto
- School of Information and Data Sciences, Nagasaki University, 1-14, Bunkyo-Machi, Nagasaki City, Nagasaki, 852-8521, Japan
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics, Wako, Saitama, 351-0198, Japan
| | - Haruka Ozaki
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Center for Artificial Intelligence Research, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|