1
|
Xing Y, Lin X. Challenges and advances in the management of inflammation in atherosclerosis. J Adv Res 2025; 71:317-335. [PMID: 38909884 DOI: 10.1016/j.jare.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 06/25/2024] Open
Abstract
INTRODUCTION Atherosclerosis, traditionally considered a lipid-related disease, is now understood as a chronic inflammatory condition with significant global health implications. OBJECTIVES This review aims to delve into the complex interactions among immune cells, cytokines, and the inflammatory cascade in atherosclerosis, shedding light on how these elements influence both the initiation and progression of the disease. METHODS This review draws on recent clinical research to elucidate the roles of key immune cells, macrophages, T cells, endothelial cells, and clonal hematopoiesis in atherosclerosis development. It focuses on how these cells and process contribute to disease initiation and progression, particularly through inflammation-driven processes that lead to plaque formation and stabilization. Macrophages ingest oxidized low-density lipoprotein (oxLDL), which partially converts to high-density lipoprotein (HDL) or accumulates as lipid droplets, forming foam cells crucial for plaque stability. Additionally, macrophages exhibit diverse phenotypes within plaques, with pro-inflammatory types predominating and others specializing in debris clearance at rupture sites. The involvement of CD4+ T and CD8+ T cells in these processes promotes inflammatory macrophage states, suppresses vascular smooth muscle cell proliferation, and enhances plaque instability. RESULTS The nuanced roles of macrophages, T cells, and the related immune cells within the atherosclerotic microenvironment are explored, revealing insights into the cellular and molecular pathways that fuel inflammation. This review also addresses recent advancements in imaging and biomarker technology that enhance our understanding of disease progression. Moreover, it points out the limitations of current treatment and highlights the potential of emerging anti-inflammatory strategies, including clinical trials for agents such as p38MAPK, tumor necrosis factor α (TNF-α), and IL-1β, their preliminary outcomes, and the promising effects of canakinumab, colchicine, and IL-6R antagonists. CONCLUSION This review explores cutting-edge anti-inflammatory interventions, their potential efficacy in preventing and alleviating atherosclerosis, and the role of nanotechnology in delivering drugs more effectively and safely.
Collapse
Affiliation(s)
- Yiming Xing
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China
| | - Xianhe Lin
- Cardiology Department, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, 230022, China.
| |
Collapse
|
2
|
Sun L, Yin Y, Cao Y, Chen C, Guo Y, Cai Z, Wu J, Li Q. Proteomic analysis of B cells in peripheral lymphatic system reveals the dynamics during the systemic lupus erythematosus progression. BIOPHYSICS REPORTS 2025; 11:129-142. [PMID: 40308935 PMCID: PMC12035744 DOI: 10.52601/bpr.2024.240045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/09/2024] [Indexed: 05/02/2025] Open
Abstract
In this study, we conducted a comprehensive proteomic analysis of B cells from the spleen, mesenteric lymph nodes (mLN), and peripheral blood mononuclear cells (PBMC) in a time-course model of systemic lupus erythematosus (SLE) using female MRL/lpr mice. By combining fluorescence-activated cell sorting (FACS) and 4D-Data-Independent Acquisition (4D-DIA) mass spectrometry, we quantified nearly 8000 proteins, identifying significant temporal and tissue-specific proteomic changes during SLE progression. PBMC-derived B cells exhibited early proteomic alterations by Week 9, while spleen-derived B cells showed similar changes by Week 12. We identified key regulatory proteins, including BAFF, BAFFR, and NFKB2, involved in B cell survival and activation, as well as novel markers such as CD11c and CD117, which have previously been associated with other immune cells. The study highlights the dynamic reprogramming of B cell proteomes across different tissues, with distinct contributions to SLE pathogenesis, providing valuable insights into the molecular mechanisms underlying B cell dysregulation in lupus. These findings offer potential therapeutic targets and biomarkers for SLE.
Collapse
Affiliation(s)
- Liming Sun
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yuanyuan Yin
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yuqing Cao
- School of Life Science and Technology, ShanghaiTech university, Shanghai 201210, China
| | - Chunlei Chen
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yutong Guo
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zeming Cai
- School of Life Science and Technology, ShanghaiTech university, Shanghai 201210, China
| | - Jiarui Wu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Qingrun Li
- CAS Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
3
|
Lu B, Chen Q, Liao X, Luo Q. Therapeutic progress in the targeting of B cells in lupus nephritis: pathogenesis to clinical research. Int Urol Nephrol 2025:10.1007/s11255-025-04441-1. [PMID: 40299182 DOI: 10.1007/s11255-025-04441-1] [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: 01/07/2025] [Accepted: 02/23/2025] [Indexed: 04/30/2025]
Abstract
Lupus nephritis (LN) is a common complication in patients with systemic lupus erythematosus (SLE), where the key mechanism is the deposition of immune complexes in the kidneys, leading to renal damage. B lymphocytes play a crucial role in the pathogenesis of lupus nephritis through several pathways. These include the production of autoantibodies, which contribute to the deposition of immune complexes in the kidneys, activation of the complement system, and promotion of local inflammatory responses. Additionally, B cells act as antigen-presenting cells, facilitating T cell activation, and secrete proinflammatory cytokines that further exacerbate inflammation. Moreover, an imbalance in B cell subpopulations can worsen autoimmune damage, highlighting the complex role of B cells in the progression of LN. Targeting B cells has emerged as a promising therapeutic strategy, particularly for patients with relapsed or refractory LN. Recent advances in B cell-targeted therapies have shown significant clinical potential, offering new hope for better disease management. This review highlights the latest progress in B cell-targeted approaches for LN treatment and explores their potential to revolutionize care for this challenging condition.
Collapse
Affiliation(s)
- Beibei Lu
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qingsong Chen
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Xiaohui Liao
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Qian Luo
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
4
|
Matsubara T, Takita M, Sekai I, Omaru N, Okai N, Morita M, Kamata K, Minaga K, Watanabe T, Kudo M. Development of Diffuse Large B Cell Lymphoma in a Patient with Systemic Lupus Erythematosus within Two Years after the Initiation of Anifrolumab and Mycophenolate Mofetil Treatment. Intern Med 2025:5362-25. [PMID: 40222946 DOI: 10.2169/internalmedicine.5362-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025] Open
Abstract
Anifrolumab is a newly developed biological agent for patients with moderate-to-severe systemic lupus erythematosus (SLE). Anifrolumab neutralizes the signaling pathways mediated by type I interferons (IFNs), which are involved in viral clearance and anticancer immunity. Although susceptibility to viral infections has been reported in patients treated with anifrolumab, whether anifrolumab treatment increases the cancer risk in patients with SLE is unknown. We herin report a case of SLE that manifested as aggressive diffuse large B-cell lymphoma (DLBCL) after treatment with anifrolumab. The neutralization of type I IFNs by anifrolumab may promote the development of DLBCL owing to defective anticancer immunity.
Collapse
Affiliation(s)
- Takuya Matsubara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Masahiro Takita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Ikue Sekai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Naoya Omaru
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Natsuki Okai
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Masahiro Morita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Ken Kamata
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Japan
| |
Collapse
|
5
|
La Gualana F, Olivieri G, Petriti B, Picciariello L, Natalucci F, Sciannamea M, Gragnani L, Basile U, Casato M, Spinelli FR, Stefanini L, Basili S, Visentini M, Ceccarelli F, Conti F. Early decrease of T-bet + B cells during subcutaneous belimumab predicts response to therapy in systemic lupus erythematosus patients. Immunol Lett 2025; 272:106962. [PMID: 39643119 DOI: 10.1016/j.imlet.2024.106962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Systemic lupus erythematosus (SLE) is characterized by B cell dysregulation and expansion of atypical B cells that may correlate with disease manifestations and activity. This study investigated the impact of subcutaneous (sc) Belimumab (BLM) on the peripheral B cell compartment and on the functional properties of CD21low, T-bet+ and CD11c+ atypical B cells, in 21 active SLE patients over a 12-month period. At baseline, active SLE patients displayed reduced unswitched IgM memory B cells and expansion of atypical B cells, compared to healthy donors and to SLE patients in remission. sc BLM therapy promptly restored B cell homeostasis with a reduction of T-bet+ B cells, observed early in patients responsive to therapy. These findings highlight the pathogenic role of T-bet+ B cells in SLE disease and suggest their potential utility as biomarker of clinical response.
Collapse
Affiliation(s)
- Francesca La Gualana
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Giulio Olivieri
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Begi Petriti
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Licia Picciariello
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Francesco Natalucci
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Maddalena Sciannamea
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Laura Gragnani
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Pisa, Italy
| | - Umberto Basile
- Department of Clinical Pathology, Santa Maria Goretti HospitalAUSL Latina, Latina, Italy
| | - Milvia Casato
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Francesca Romana Spinelli
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Lucia Stefanini
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Stefania Basili
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Marcella Visentini
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy.
| | - Fulvia Ceccarelli
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| | - Fabrizio Conti
- Lupus Clinic, Rheumatology, Dipartimento di Scienze Cliniche Internistiche Anestesiologiche e Cardiovascolari, Sapienza Università di Roma, Viale del Policlinico 155 00161 Rome, Italy
| |
Collapse
|
6
|
Wang Y, Xu Y. Advances in maintenance therapies for neuromyelitis optica spectrum disorders: A new era of targeted drugs. Mult Scler Relat Disord 2025; 96:106351. [PMID: 40036907 DOI: 10.1016/j.msard.2025.106351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Neuromyelitis optica spectrum disorder (NMOSD) is a rare inflammatory demyelinating disease of the central nervous system, mainly affecting the optic nerve and spinal cord, with a high rate of recurrence and disability. The spectrum of possibilities for NMOSD maintenance therapies has increased recently due to the development of novel medications based on a deeper understanding of its pathogenesis. METHODS Clinical and preclinical advances in NMOSD maintenance therapies were searched in PubMed up until July 20, 2024. RESULTS We reviewed the treatment options including approved monoclonal antibodies, clinical trial drugs, and preclinical targets. Both randomized clinical trials and real-word studies have evaluated the safety and efficacy of monoclonal antibodies against complement, IL-6 receptor and B cells. Drugs in the early stage of clinical trials are mainly focused on small molecule targeted drugs and cell-based therapies. Additionally, preclinical studies aimed to explore new targets, such as aquaporin blocking antibodies, based on experiences in cell cultures and animal models. CONCLUSIONS From biologics to small molecules and cells, researchers have endeavored to provide better medical care for NMOSD patients. With the emergence of more and more targeted drugs, it is anticipated that the treatment of NMOSD will enter a new era.
Collapse
Affiliation(s)
- Yingjie Wang
- Center of Multiple Sclerosis and Related Disorders, China; Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| | - Yan Xu
- Center of Multiple Sclerosis and Related Disorders, China; Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Jiang K, Pan Y, Pu D, Shi L, Xu X, Bai M, Gong X, Guo J, Li M. Kidney transplantation in Lupus Nephritis: a comprehensive review of challenges and strategies. BMC Surg 2025; 25:112. [PMID: 40121458 PMCID: PMC11929324 DOI: 10.1186/s12893-025-02832-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
PURPOSE OF REVIEW: Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus (SLE), significantly impacting patient outcomes. Despite advances in immunosuppressive therapies, many patients progress to end-stage renal disease (ESRD), and kidney transplantation becomes essential for improving survival. However, the unique characteristics of autoimmune diseases make the timing of kidney transplantation and post-transplant management challenging. This review evaluates authoritative guidelines and recent studies to identify optimal timing for kidney transplantation and effective pre- and post-transplant management measures for patients with LN. RECENT FINDINGS: Advancements in immunosuppressive therapies, including calcineurin inhibitors, Voclosporin, and biologic agents such as belimumab, have significantly improved LN management. Emerging biomarkers, such as urinary MCP-1 and BAFF, offer promising tools for monitoring LN activity and predicting recurrence risk post-transplantation. Current guidelines emphasize the importance of achieving disease quiescence before transplantation, while new evidence supports the benefits of preemptive transplantation and personalized immunosuppressive regimens in improving patient and graft survival. This review highlights the latest evidence and strategies for optimizing kidney transplantation outcomes in LN patients, focusing on timing, immunosuppression, and disease monitoring.
Collapse
Affiliation(s)
- Kerong Jiang
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China.
| | - Yongsheng Pan
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Dan Pu
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Lijuan Shi
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Xiaoliang Xu
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Minfeng Bai
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Xiaqiong Gong
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Jie Guo
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| | - Ming Li
- Department of Clinical Laboratory, The First People's Hospital of Kunming, No. 1228 Peking Road, Kunming, 650051, China
| |
Collapse
|
8
|
Tan J, Huang H, Tan L, Li B. Telitacicept for systemic lupus erythematosus with post‑surgical papillary thyroid carcinoma: A case report. Biomed Rep 2025; 22:48. [PMID: 39882332 PMCID: PMC11775643 DOI: 10.3892/br.2025.1926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/05/2024] [Indexed: 01/31/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with a complex etiology primarily linked to abnormalities in B lymphocytes within the human body, resulting in the production of numerous pathogenic autoantibodies. Telitacicept is a relatively novel humanized, recombinant transmembrane activator, calcium modulator and cyclophilin ligand interactor fused with the Fc portion (TACI-Fc). It works by competitively inhibiting the TACI site, neutralizing the activity of B-cell lymphocyte stimulator and A proliferation-inducing ligand. This, in turn, inhibits the development and survival of plasma cells and mature B cells. A 28-year-old female was admitted to the Department of Rheumatology and Immunology (People's Hospital of Longhua; Shenzhen, China) in June 2021 due to systemic edema for more than a month and hair loss lasting for a week. After comprehensive examination, the patient was diagnosed with SLE with hematological system involvement, serositis, lupus nephritis and secondary antiphospholipid syndrome. After receiving medications including glucocorticoids, mycophenolate mofetil and cyclosporine, the patient's white blood cells, platelets, hemoglobin, urinary protein and multiple serositis returned to normal. However, the levels of complement 3 (C3) and C4 did not significantly improve. Subsequently, the patient underwent thyroid ultrasound examination, which suggested thyroid nodules. After thyroid puncture biopsy, the patient was diagnosed with papillary thyroid carcinoma (PTC). After surgical resection, the patient was confirmed to have PTC by pathological biopsy, with no lymph node metastasis. At two months after surgery, the patient was treated with telitacicept, and the complement levels not only returned to normal but also remained stable for a long time. The present case was the first to report the use of telitacicept for the successful treatment of a patient with SLE with post-surgical PTC, providing a potential therapeutic option for SLE with a prior history of carcinoma. The role of telitacept in this field requires further research and attention.
Collapse
Affiliation(s)
- Jinhui Tan
- Department of Rheumatology and Immunology, People's Hospital of Longhua, Shenzhen, Guangdong 518109, P.R. China
| | - Hai Huang
- Department of Health Management, People's Hospital of Longhua, Shenzhen, Guangdong 518109, P.R. China
| | - Linghua Tan
- Department of Health Management, Jiangmen Wuyi Hospital of Chinese Medicine, Jiangmen, Guangdong 529099, P.R. China
| | - Bo Li
- Department of Rheumatology and Immunology, People's Hospital of Longhua, Shenzhen, Guangdong 518109, P.R. China
| |
Collapse
|
9
|
Iwasaki T, Yoshifuji H, Kitagori K, Sumitomo S, Akizuki S, Nakashima R, Tsuji H, Hiwa R, Shirakashi M, Murakami K, Onishi A, Onizawa H, Tanaka M, Matsuda F, Morinobu A, Ohmura K. Memory B cells and their transcriptomic profiles associated with belimumab resistance in systemic lupus erythematosus in the maintenance phase. Front Immunol 2025; 16:1506298. [PMID: 39975549 PMCID: PMC11835923 DOI: 10.3389/fimmu.2025.1506298] [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: 10/04/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
The factors contributing to the treatment efficacy of belimumab in patients with systemic lupus erythematosus (SLE) in the maintenance phase are unknown. Here, we collected blood samples from patients with SLE (n=44) treated with belimumab before and three and six months after treatment. RNA-Seq of whole blood was performed, and gene expression was quantified. Immune cell type enrichment analysis estimated immune cell subtype proportions and gene expression in each subtype. The Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K) < 4 at six months was set as the primary efficacy criterion. Non-responders exhibited upregulated B cell proliferation signals before treatment, associated with an increased number of memory B cells. A higher proportion of memory B cells before treatment predicted poor response (p=5.1×10-4). This was also associated with changes in disease activity and glucocorticoid dose at six months compared with baseline. Belimumab did not affect memory B cell proportion during the treatment time course, in contrast to naïve B cells. Higher memory B cell proportion was associated with higher type-I interferon (IFN) scores and lower white blood cell and complement C4 levels. Transcriptomic analysis of memory B cells in non-responders revealed significant upregulation of immunoglobulin genes (Ig). Memory B cells and high Ig expression in them were identified as a treatment-resistant factor of belimumab in SLE patients. Lower C4 and white blood cell counts may serve as clinical markers of higher memory B cells.
Collapse
Affiliation(s)
- Takeshi Iwasaki
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
- Graduate School of Medicine, Kyoto University, Center for Genomic Medicine, Kyoto, Japan
| | - Hajime Yoshifuji
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Koji Kitagori
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Shuji Sumitomo
- Kobe City Medical Center General Hospital, Department of Rheumatology, Kobe, Japan
| | - Shuji Akizuki
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Ran Nakashima
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Hideaki Tsuji
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Ryosuke Hiwa
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Mirei Shirakashi
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Kosaku Murakami
- Division of Clinical Immunology and Cancer Immunotherapy, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Onishi
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideo Onizawa
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Graduate School of Medicine, Kyoto University, Center for Genomic Medicine, Kyoto, Japan
| | - Akio Morinobu
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
| | - Koichiro Ohmura
- Graduate School of Medicine, Kyoto University, Department of Rheumatology and Clinical Immunology, Kyoto, Japan
- Kobe City Medical Center General Hospital, Department of Rheumatology, Kobe, Japan
| |
Collapse
|
10
|
Uppin V, Gibbons H, Troje M, Feinberg D, Webber BR, Moriarity BS, Parameswaran R. CAR-T cell targeting three receptors on autoreactive B cells for systemic lupus erythematosus therapy. J Autoimmun 2025; 151:103369. [PMID: 39832454 DOI: 10.1016/j.jaut.2025.103369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated B cell activation, autoantibody production, and nephritis. B cell activating factor (BAFF) overexpression enhances autoreactive B-cell survival, driving autoimmunity. BAFF specific belimumab and CD20 specific rituximab antibodies are used for SLE therapy but are not curative, highlighting the need for alternative B cell depletion therapies. Here, we use BAFF ligand based chimeric antigen receptor T (CAR-T) cells targeting BAFFr, BCMA and TACI expressed on mature B cells and plasma cells. BAFF CAR-T cells efficiently killed B cells after co-culture with peripheral blood mononuclear cells (PBMCs) from SLE patients and in a patient derived SLE xenograft humanized mouse model developed by injecting patient PBMCs into immunocompromised mice. We also generated murine CD8+ T cells expressing human BAFF CAR to test their therapeutic efficacy in spontaneous (MRL/lpr) and pristane induced mouse models of SLE. In both models, BAFF CAR-T cells mediated persistent elimination of mature B cells, resulting in a decrease in the production of autoantibodies (IgM, IgG, Anti-ANA, and Anti-dsDNA IgG) and proteinuria along with prolonged survival. Adoptive transfer of B cells from control MRL/lpr lupus mice to previously BAFF CAR-T treated MRL/lpr lupus mice showed continued depletion of B cells and prolonged survival. Potential advantages of BAFF CAR-T therapy include avoiding B cell aplasia as BAFF receptors are not expressed by early B cells and preventing the escape of long-lived plasma cells post BAFF CAR-T therapy as they express receptors of BAFF. These data demonstrate the potential for a cellular immunotherapy based approach to induce remission of SLE pathogenesis using BAFF-CAR-T therapy.
Collapse
Affiliation(s)
- Vinayak Uppin
- Division of Haematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Daniel Feinberg
- Division of Haematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | | - Reshmi Parameswaran
- Division of Haematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA; Pediatric Haematology and Oncology, The Angie Fowler Adolescent & Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH, USA; The Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
11
|
Dedemadi AG, Gkolfinopoulou C, Nikoleri D, Nikoloudaki M, Ruhanen H, Holopainen M, Käkelä R, Christopoulou G, Bournazos S, Constantoulakis P, Sidiropoulos P, Bertsias G, Chroni A. Improvement of high-density lipoprotein atheroprotective properties in patients with systemic lupus erythematosus after belimumab treatment. Rheumatology (Oxford) 2025; 64:648-657. [PMID: 38514392 PMCID: PMC11781589 DOI: 10.1093/rheumatology/keae192] [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: 01/07/2024] [Revised: 03/14/2024] [Accepted: 03/16/2024] [Indexed: 03/23/2024] Open
Abstract
OBJECTIVE Chronic inflammatory diseases, like Systemic Lupus Erythematosus (SLE), carry an increased risk for atherosclerosis and cardiovascular events, accompanied by impairment of atheroprotective properties of high-density lipoprotein (HDL). In SLE, serum B cell-activating factor (BAFF), a cytokine implicated in disease progression, has been correlated with subclinical atherosclerosis. We investigated the impact of treatment with belimumab -an anti-BAFF monoclonal antibody- on HDL atheroprotective properties and composition in SLE patients. METHODS Serum samples were collected from 35 SLE patients with active disease despite conventional therapy, before and after 6-month add-on treatment with belimumab, and 26 matched healthy individuals. We measured cholesterol efflux and antioxidant capacities, paraoxonase-1 (PON1) activity, serum amyloid A1 (SAA1), myeloperoxidase (MPO) and lipid peroxidation product levels of HDL. LC-MS/MS was performed to analyse the HDL lipidome. RESULTS Following treatment with belimumab, cholesterol efflux and antioxidant capacities of HDL were significantly increased in SLE patients and restored to levels of control subjects. HDL-associated PON1 activity was also increased, whereas lipid peroxidation products were decreased following treatment. HDL cholesterol efflux and antioxidant capacities correlated negatively with the disease activity. Changes were noted in the HDL lipidome of SLE patients following belimumab treatment, as well as between SLE patients and healthy individuals, and specific changes in lipid species correlated with functional parameters of HDL. CONCLUSIONS HDL of SLE patients with active disease displays impaired atheroprotective properties accompanied by distinct lipidomic signatures compared with controls. Belimumab treatment may improve the HDL atheroprotective properties and modify the HDL lipidomic signature in SLE patients, thus potentially mitigating atherosclerosis development.
Collapse
Affiliation(s)
- Anastasia-Georgia Dedemadi
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, Athens, Greece
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Athens, Greece
| | - Christina Gkolfinopoulou
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, Athens, Greece
| | - Dimitra Nikoleri
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Myrto Nikoloudaki
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
| | - Hanna Ruhanen
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, and Biocenter Finland, Helsinki, Finland
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Minna Holopainen
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, and Biocenter Finland, Helsinki, Finland
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, HiLIPID, Helsinki Institute of Life Science, HiLIFE, and Biocenter Finland, Helsinki, Finland
- Molecular and Integrative Biosciences Research Program, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | | | | | | | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - George Bertsias
- Laboratory of Rheumatology, Autoimmunity and Inflammation, University of Crete Medical School, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research “Demokritos”, Agia Paraskevi, Athens, Greece
| |
Collapse
|
12
|
Jägerback S, Gomez A, Parodis I. Predictors of renal flares in systemic lupus erythematosus: a post-hoc analysis of four phase III clinical trials of belimumab. Rheumatology (Oxford) 2025; 64:623-631. [PMID: 38216728 PMCID: PMC11781576 DOI: 10.1093/rheumatology/keae023] [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: 11/29/2023] [Accepted: 12/04/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE The objective of this study was to identify predictors of renal flares in patients with SLE treated for active extra-renal disease. METHODS Data from four clinical trials of belimumab in SLE (BLISS-52, NCT00424476; BLISS-76, NCT00410384; BLISS-NEA, NCT01345253; BLISS-SC, NCT01484496) were used. Patients were assigned to belimumab or placebo on top of standard therapy. We investigated the performance of predictors of renal flares through weeks 52-76 using proportional hazards regression analysis. RESULTS Of 3225 participants, 192 developed at least one renal flare during follow-up, with the first occurring after a median time of 197 days. Current/former renal involvement [hazards ratio (HR): 15.4; 95% CI: 8.3-28.2; P < 0.001], low serum albumin levels (HR 0.9; 95% CI: 0.8-0.9; P < 0.001), proteinuria (HR: 1.6; 95% CI: 1.5-1.7; P < 0.001), and low C3 levels (HR: 2.9; 95% CI: 2.1-4.1; P < 0.001) at baseline appeared robust determinants of impending renal flares. Anti-dsDNA positivity yielded an increased hazard for renal flares (HR: 2.1; 95% CI: 1.4-3.2; P < 0.001), which attenuated after adjustments. Anti-Sm positivity was associated with renal flares in the placebo (HR: 3.7; 95% CI: 2.0-6.9; P < 0.001) but not in the belimumab subgroup, whereas anti-ribosomal P positivity was associated with renal flares in the belimumab subgroup only (HR: 2.8; 95% CI: 1.5-5.0; P = 0.001). CONCLUSION A history of renal involvement, high baseline proteinuria, hypoalbuminaemia, and C3 consumption were robust determinants of impending renal flares. In addition to anti-dsDNA, anti-Sm and anti-ribosomal P protein antibody positivity may have value in surveillance of renal SLE.
Collapse
Affiliation(s)
- Sandra Jägerback
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine, Danderyd University Hospital, Danderyd, Sweden
| | - Alvaro Gomez
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet and 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
|
13
|
Yang Z, Wu Y, Huang S, Bao J, Xu L, Fan Y. Risk factors associated with thrombocytopenia in systemic lupus erythematosus: A systematic review and meta-analysis. Autoimmun Rev 2025; 24:103721. [PMID: 39667603 DOI: 10.1016/j.autrev.2024.103721] [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: 10/14/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) frequently manifests with thrombocytopenia (TP), a hematologic complication that heightens the risk of severe outcomes and increases mortality. This meta-analysis aims to evaluate the potential risk factors associated with TP in SLE patients, providing insights into the demographic features, clinical features, and laboratory findings that contribute to this condition. METHODS A comprehensive literature search was conducted across eight databases from inception to September 1, 2024. Study quality was assessed using the Newcastle-Ottawa Scale. Meta-analysis was conducted using univariate and multivariate analyses with Revman 5.3, while heterogeneity was addressed through subgroup and sensitivity analyses. Publication bias was assessed using funnel plots and Egger tests via Stata 15.0. RESULTS Seventeen high-quality studies meeting the inclusion criteria were incorporated into this meta-analysis. Independent risk factors for TP in SLE included age (Demographic Features), serositis, splenomegaly, blood system involvement, and renal involvement (Clinical Features), as well as cardiac involvement, anemia, leukocytopenia, low C3/C4, ACA, and CRP (Laboratory Findings). Arthritis and rash were protective factors. Subgroup analysis addressed heterogeneity caused by unit and sample size differences. Sensitivity analysis comparing the consistency between fixed-effects model (FEM) and random-effects model (REM) confirmed the reliability of the findings, and both funnel plots and Egger tests suggested no publication bias. CONCLUSION This meta-analysis identified several potential independent risk factors for TP in SLE. Early screening and timely intervention for patients with these risk factors are essential to reduce the likelihood of TP, prevent severe organ damage, and improve overall prognosis.
Collapse
Affiliation(s)
- Ze Yang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yanzuo Wu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Shuo Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Jie Bao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Li Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| | - Yongsheng Fan
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310005, China.
| |
Collapse
|
14
|
Tapryal S. Monoclonal antibodies - A repertoire of therapeutics. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 144:151-212. [PMID: 39978966 DOI: 10.1016/bs.apcsb.2024.11.001] [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: 02/22/2025]
Abstract
Antibodies are a class of biomolecules armed with extraordinary diversity, unmatched in the biological world by any other class of molecules. This characteristic feature equips antibodies to recognize, bind, and eliminate an infinite number of pathogens/antigens facilitated by their effector functions. The repertoire of natural binding specificities of antibodies (Abs) is greater than the calculated estimate of ∼1012 in humans, as a naive, single antigen-binding site may bind more than one antigen employing the plasticity in antigen-antibody interactions, potentiating Abs to fight infinite pathogenic insults and restrict the development of cancers. Additionally, advanced technological interventions, by allowing manipulation of the germline and acquired specificities of human/animal immunoglobulins (Ig) have contributed immensely to broaden their existing repertoire and scope of clinical applications. The available natural repertoire of Ig and Ig-like molecules in other animals, e.g., mice, horses, cows, pigs, rabbits, camels, llamas, etc., further diversified the source of unique antigen-binding specificities. The recombinant DNA technology, in association with hybridoma , transgenic, and phage display technologies, has helped create a parallel repertoire of unique antibody molecules [animal Abs, camelid heavy chain Abs (hcAbs), chimeric Abs, chimeric hcAbs, humanized Abs, humanized nanobody (Nb)-hcAbs, human Abs, etc.], monoclonal Ab (mAb) derived fragments [antigen-binding-fragment (Fab), single-chain-variable-fragment (scFv), variable-fragement (Fv), single-variable-domain of hcAbs (VHH), bispecific scFv, diabodies, triabodies, intrabodies, bispecific Fabs, tri-specific Fabs, etc.), and immunoconjugates generated by fusing/conjugating mAb fragments with enzyme, toxin, prodrug etc., molecules. The current chapter provides a detailed description of the natural and engineered antibody repertoires and discusses various strategies using which these molecules are being inducted as novel immunotherapeutics for treating a significant number of human diseases.
Collapse
Affiliation(s)
- Suman Tapryal
- Department of Biophysics, University of Delhi, South Campus, Benito Juarez Road, South Moti Bagh, New Delhi, India.
| |
Collapse
|
15
|
Wang X, Gao M, Song J, Li M, Chen Y, Lv Y, Jia W, Wan B. Differential Expression of tRNA-Derived Small RNA Markers of Antidepressant Response and Functional Forecast of Duloxetine in MDD Patients. Genes (Basel) 2025; 16:162. [PMID: 40004491 PMCID: PMC11855652 DOI: 10.3390/genes16020162] [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: 11/30/2024] [Revised: 01/14/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Duloxetine, despite being a leading treatment option for major depressive disorder (MDD), exhibits a relatively low adequate response rate when used as a monotherapy, and the fundamental molecular mechanisms remain largely elusive. tRNA-derived small RNA (tsRNA) is a particularly interesting and new class of molecules that is becoming increasingly noticeable for investigation. METHODS We integrated small RNA sequencing with bioinformatics approaches to dissect the expression profiles of tsRNAs and decipher their functional roles post-duloxetine treatment. Subsequently, molecular docking experiments were carried out to validate the potential functions. RESULTS Ten tsRNAs significantly changed in the duloxetine response group after an 8-week therapy. Correlation analyses revealed that these tsRNAs predominantly interacted with miRNAs across multiple biological pathways and processes, such as the ECM-receptor interaction and B cell activation. Molecular docking analysis corroborated the binding capabilities of duloxetine with key proteins associated with ECM1 and BAFF, respectively. CONCLUSIONS The identified changes in tsRNAs can precisely mirror the response of duloxetine in MDD treatment, offering novel insights into the underlying mechanisms of duloxetine action.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.)
| | - Ming Gao
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jing Song
- School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Miaolong Li
- School of Chemical Science and Engineering, Tongji University, Shanghai 200070, China
| | - Yu Chen
- Department of Clinical Medicine, He University, Shenyang 110163, China
| | - Yingfang Lv
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.)
| | - Wei Jia
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Bingbing Wan
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China; (X.W.)
| |
Collapse
|
16
|
Wang Y, Zhao R, Liang Q, Ni S, Yang M, Qiu L, Ji J, Gu Z, Dong C. Organ-based characterization of B cells in patients with systemic lupus erythematosus. Front Immunol 2025; 16:1509033. [PMID: 39917309 PMCID: PMC11798990 DOI: 10.3389/fimmu.2025.1509033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic, inflammatory, and progressive autoimmune disease. The unclear pathogenesis, high heterogeneity, and prolonged course of the disease present significant challenges for effective clinical management of lupus patients. Dysregulation of the immune system and disruption of immune tolerance, particularly through the abnormal activation of B lymphocytes and the production of excessive autoantibodies, lead to widespread inflammation and tissue damage, resulting in multi-organ impairment. Currently, there is no systematic review that examines the specificity of B cell characteristics and pathogenic mechanisms across various organs. This paper reviews current research on B cells in lupus patients and summarizes the distinct characteristics of B cells in different organs. By integrating clinical manifestations of organ damage in patients with a focus on the organ-specific features of B cells, we provide a new perspective on enhancing the efficacy of lupus-targeted B cell therapy strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| | - Chen Dong
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Clinical Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, China
| |
Collapse
|
17
|
Diehl C, Soberón V, Baygün S, Chu Y, Mandelbaum J, Kraus L, Engleitner T, Rudelius M, Fangazio M, Daniel C, Bortoluzzi S, Helmrath S, Singroul P, Gölling V, Osorio Barrios F, Seyhan G, Oßwald L, Kober-Hasslacher M, Zeng T, Öllinger R, Afzali AM, Korn T, Honarpisheh M, Lech M, Ul Ain Q, Pircher J, Imširović V, Jelenčić V, Wensveen FM, Passerini V, Bärthel S, Bhagat G, Dominguez-Sola D, Saur D, Steiger K, Rad R, Pasqualucci L, Weigert O, Schmidt-Supprian M. Hyperreactive B cells instruct their elimination by T cells to curb autoinflammation and lymphomagenesis. Immunity 2025; 58:124-142.e15. [PMID: 39729992 DOI: 10.1016/j.immuni.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/14/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024]
Abstract
B cell immunity carries the inherent risk of deviating into autoimmunity and malignancy, which are both strongly associated with genetic variants or alterations that increase immune signaling. Here, we investigated the interplay of autoimmunity and lymphoma risk factors centered around the archetypal negative immune regulator TNFAIP3/A20 in mice. Counterintuitively, B cells with moderately elevated sensitivity to stimulation caused fatal autoimmune pathology, while those with high sensitivity did not. We resolved this apparent paradox by identifying a rheostat-like cytotoxic T cell checkpoint. Cytotoxicity was instructed by and directed against B cells with high intrinsic hyperresponsiveness, while less reactive cells were spared. Removing T cell control restored a linear relationship between intrinsic B cell reactivity and lethal lymphoproliferation, lymphomagenesis, and autoinflammation. We thus identify powerful T cell-mediated negative feedback control of inherited and acquired B cell pathogenicity and define a permissive window for autoimmunity to emerge.
Collapse
Affiliation(s)
- Carina Diehl
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Valeria Soberón
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Seren Baygün
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Yuanyuan Chu
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Jonathan Mandelbaum
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Laura Kraus
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thomas Engleitner
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Marco Fangazio
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Christoph Daniel
- Department of Nephropathology, Faculty of Medicine, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Sabrina Bortoluzzi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Sabine Helmrath
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Pankaj Singroul
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Vanessa Gölling
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Francisco Osorio Barrios
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Gönül Seyhan
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Lena Oßwald
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; Department of Medicine III, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Maike Kober-Hasslacher
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Theodor Zeng
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rupert Öllinger
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Ali M Afzali
- Institute for Experimental Neuroimmunology, Department of Neurology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Department of Neurology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Mohsen Honarpisheh
- Renal Division, Department of Medicine IV, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Maciej Lech
- Renal Division, Department of Medicine IV, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Qurrat Ul Ain
- Department of Medicine I, Faculty of Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Joachim Pircher
- Department of Medicine I, Faculty of Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany; Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany
| | - Vanna Imširović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Vedrana Jelenčić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Verena Passerini
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Faculty of Medicine, Department of Medicine III, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Stefanie Bärthel
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Govind Bhagat
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - David Dominguez-Sola
- Departments of Oncological Sciences and Pathology, Tisch Cancer Institute, Lipschultz Precision Immunology Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dieter Saur
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Oliver Weigert
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Faculty of Medicine, Department of Medicine III, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Marc Schmidt-Supprian
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany.
| |
Collapse
|
18
|
Gao S, Yang C, Huang B, Yang L, Lu L, Yang H, Li T, Pan Q. Comparative efficacy and safety of different recommended doses of telitacicept in patients with systemic lupus erythematosus in China: a systematic review and meta-analysis. Front Immunol 2025; 15:1472292. [PMID: 39867893 PMCID: PMC11757125 DOI: 10.3389/fimmu.2024.1472292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/19/2024] [Indexed: 01/28/2025] Open
Abstract
Background Telitacicept, a new biological agent, was approved in China for treating systemic lupus erythematosus (SLE) in 2021. Its optimal dosing for treating SLE remains unclear. Therefore, the aim of this meta-analysis is to evaluate the efficacy and safety of various telitacicept doses in SLE treatment. Methods PubMed, EMBASE, Cochrane libraries, Web of science, China National Knowledge Infrastructure (CNKI), VIP, Wanfang, and Sinomed were searched for the controlled trials that studied the efficacy and safety of telitacicept on SLE patients from their initiation to April 30, 2024. The analysis included three randomized controlled trials (RCT) with 606 participants. We used fixed-effects models for meta-analyses and the risk ratios (RRs) and corresponding 95% confidence intervals (CIs) to evaluate the effectiveness and safety. Heterogeneity was assessed and quantified using I2. Results All telitacicept dosages (80 mg, 160 mg, 240 mg) significantly improved SLE Responder Index 4 (SRI4) responses compared to the control group (RR = 2.20, 95%CI:1.50-3.21, p < 0.0001; RR = 2.18, 95%CI: 1.82-2.62, p < 0.00001; RR = 2.44, 95%CI: 1.67-3.56, p < 0.00001, respectively). The 80 mg, 160 mg, and 240 mg groups also showed better improvement on SELENA-SLE Disease Activity Index (SELENA-SLEDAI) scores (RR = 1.63, 95%CI: 1.23-2.17, p = 0.0008; RR = 1.72, 95%CI: 1.45-2.04, p < 0.00001; RR = 1.73, 95%CI: 1.30-2.30, p = 0.0002, respectively) and Physician Global Assessment (PGA) scores (RR = 1.25, 95%CI: 1.09-1.44, p = 0.002; RR = 1.39, 95%CI: 1.25-1.55, p < 0.00001; RR = 1.24, 95%CI: 1.09-1.42, p = 0.002, respectively). Furthermore, 160 mg group exhibited higher British Isles Lupus Assessment Group (BILAG) score than the control group (RR = 1.11, 95%CI: 1.01-1.22, p = 0.03). As for security, 160 mg telitacicept group had higher incidence of adverse events (AEs) than the control group (RR = 1.10, 95%CI: 1.03-1.18, p = 0.007). Conclusion Telitacicept combined with standard therapy presents potential benefits but there are certain safety concerns with certain dosages of telitacicept, warranting further investigation for optimal dosing strategies in SLE management. Systematic review registration INPLASY.COM, identifier INPLASY202440101.
Collapse
Affiliation(s)
- Shenglan Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chunlong Yang
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Bitang Huang
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lawei Yang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non−Communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lu Lu
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huiting Yang
- Clinical Research Center, Laboratory Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Qingjun Pan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Clinical Laboratory, State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
19
|
Teng Y, Xu H, He X, Zhuang Q, Lu H, Xu R, Xue D. LXRα agonist differentially regulates BAFF expression and biological effects in RAW264.7 cells depending on growth status: LXRα activation and BAFF signaling in RAW264.7 cells. Biochem Biophys Res Commun 2025; 742:151067. [PMID: 39632295 DOI: 10.1016/j.bbrc.2024.151067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
B- cell-activating factor (BAFF), which is essential for the survival and development of B cells, is mainly produced by myeloid cells such as macrophages. Abnormal macrophage infiltration and high BAFF expression in kidney allografts are associated with the occurrence and development of antibody-mediated rejection (ABMR). Nuclear hormone receptor Liver X receptors (LXRs), is a nonnegligible participant in regulating cholesterol metabolism and inflammatory responses. Nowadays the effects of LXRα activation on macrophages have been widely studied, however the effects of LXRα activation on BAFF expression and cell function due to the change of BAFF signaling have not yet been fully investigated. In the present study, LXRα activation alone was found to downregulate BAFF expression in quiescent RAW 264.7 cells, whereas LXRα agonist significantly upregulated BAFF expression in cells pretreated with lipopolysaccharide (LPS) for 6 h. The increased BAFF signaling promoted M1 polarization and enhanced cell viability, migration, and phagocytic ability. LXRα can directly bind to the BAFF promoter region and decrease BAFF expression in RAW264.7 cells. LXRα activation enhanced mitochondrial metabolism, which promoted BAFF expression in the LPS-activated cells. Our results indicate that subtle changes in the microenvironment would affect the biological function of macrophages, in which a variety of BAFF signaling pathways may also be involved, providing a new perspective on exploring the mechanism of allograft rejection and uncovering the potential reason for the unstable efficacy of anti-BAFF preparations in kidney transplant recipients.
Collapse
Affiliation(s)
- Yisa Teng
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Haiyan Xu
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Xiaozhou He
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qianfeng Zhuang
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Lu
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Renfang Xu
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Xue
- Urology Department, Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
20
|
Zhao Y, Qi F, Bai J, Zhang N, Yang T, Sun W, Li X, Wei W. Real-world efficacy of belimumab in systemic lupus erythematosus: a prospective cohort from a single centre in China. Rheumatology (Oxford) 2025; 64:108-116. [PMID: 38011668 DOI: 10.1093/rheumatology/kead629] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVE The objective of this study was to explore the efficacy and safety of belimumab among Chinese patients with SLE in a real-world setting. METHODS A prospective cohort study was performed, and SLE patients taking belimumab on a background of standard-of-care (SoC) treatment were consecutively enrolled from July 2021 to December 2022. Based on baseline characteristics, the patients were divided into three groups: the newly diagnosed group, the relapsed group and the refractory group. Patients in the newly diagnosed group were newly diagnosed with SLE within 4 weeks of starting belimumab. Patients in the relapse group had experienced a severe flare. Refractory patients were patients with unsatisfactory GC taper and/or disease activity control. Clinical data were collected, and disease assessments were conducted regularly. Newly diagnosed patients with SoC alone and healthy controls (HCs) were also enrolled. RESULTS A total of 123 SLE patients were included in the analysis, with a median follow-up period of 12 months (range 3-18 months). Thirty-three out of 123 patients were newly diagnosed, 32 had relapsed disease, and 58 had refractory disease. The SLE Responder Index 4 (SRI-4) response was achieved with good tolerance by 55.77% of patients at 3 months, 56.63% at 6 months, 63.24% at 9 months, 63.64% at 12 months and 57.14% at 18 months. Serological parameters (anti-dsDNA and C3/C4), SLEDAI-2K and daily prednisone intake were improved overall and in each group. Of the three groups, the newly diagnosed group had the highest SRI-4 rate as well as the greatest improvement in serological parameters and SLEDAI-2K. Compared with newly diagnosed patients with SoC alone, the cumulative prednisone intake of newly diagnosed patients taking belimumab was significantly decreased. CONCLUSION Our data supported the efficacy of belimumab in Chinese SLE patients in a real-life setting. Our study also provided new evidence indicating remarkable achievement of the SRI-4 response during belimumab therapy in newly diagnosed SLE patients.
Collapse
Affiliation(s)
- Yin Zhao
- Department of Nephrology and Rheumatology, The Affiliated Hospital of Yunnan University (The Second People's Hospital of Yunnan Province), Kunming, Yunnan, China
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fumin Qi
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinyu Bai
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Nephrology and Rheumatology, The Third People's Hospital of Zhengzhou, Zhengzhou, China
| | - Na Zhang
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tong Yang
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenwen Sun
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Li
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Wei
- Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
21
|
Zhou J, Lei B, Shi F, Luo X, Wu K, Xu Y, Zhang Y, Liu R, Wang H, Zhou J, He X. CAR T-cell therapy for systemic lupus erythematosus: current status and future perspectives. Front Immunol 2024; 15:1476859. [PMID: 39749335 PMCID: PMC11694027 DOI: 10.3389/fimmu.2024.1476859] [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: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Systemic lupus erythematosus (SLE) and lupus nephritis (LN) are debilitating autoimmune disorders characterized by pathological autoantibodies production and immune dysfunction, causing chronic inflammation and multi-organ damage. Despite current treatments with antimalarial drugs, glucocorticoids, immunosuppressants, and monoclonal antibodies, a definitive cure remains elusive, highlighting an urgent need for novel therapeutic strategies. Recent studies indicate that chimeric antigen receptor T-cell (CAR-T) therapy has shown promising results in treating B-cell malignancies and may offer a significant breakthrough for non-malignant conditions like SLE. In this paper, we aim to provide an in-depth analysis of the advancements in CAR-T therapy for SLE, focusing on its potential to revolutionize treatment for this complex disease. We explore the fundamental mechanisms of CAR-T cell action, the rationale for its application in SLE, and the immunological underpinnings of the disease. We also summarize clinical data on the safety and efficacy of anti-CD19 and anti-B cell maturation antigen (BCMA) CAR-T cells in targeting B-cells in SLE. We discuss the clinical implications of these findings and the potential for CAR-T therapy to improve outcomes in severe or refractory SLE cases. The integration of CAR-T therapy into the SLE treatment paradigm presents a new horizon in autoimmunity research and clinical practice. This review underscores the need for continued exploration and optimization of CAR-T strategies to address the unmet needs of SLE patients.
Collapse
Affiliation(s)
- Jincai Zhou
- Innovation & Research Department, OriCell Therapeutics Co. Ltd., Shanghai, China
| | | | | | | | | | | | | | | | | | - Joy Zhou
- Innovation & Research Department, OriCell Therapeutics Co. Ltd., Shanghai, China
| | - Xiaowen He
- Innovation & Research Department, OriCell Therapeutics Co. Ltd., Shanghai, China
| |
Collapse
|
22
|
Guo Q, Li J, Wang J, Li L, Wei J, Zhang L. The advent of chimeric antigen receptor T Cell therapy in recalibrating immune balance for rheumatic autoimmune disease treatment. Front Pharmacol 2024; 15:1502298. [PMID: 39734406 PMCID: PMC11672202 DOI: 10.3389/fphar.2024.1502298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
CAR-T cell therapy, a cutting-edge cellular immunotherapy with demonstrated efficacy in treating hematologic malignancies, also exhibits significant promise for addressing autoimmune diseases. This innovative therapeutic approach holds promise for achieving long-term remission in autoimmune diseases, potentially offering significant benefits to affected patients. Current targets under investigation for the treatment of these conditions include CD19, CD20, and BCMA, among others. However, CAR-T therapy faces difficulties such as time-consuming cell manufacturing, complex and expensive process, and the possibility of severe adverse reactions complicating the treatment, etc. This article examines CAR-T therapy across various rheumatic autoimmune diseases, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), Sjögren's syndrome (SS), systemic sclerosis (SSc), antisynthetase syndrome (ASS), and ANCA-associated vasculitis (AAV), highlighting both therapeutic advancements and ongoing challenges.
Collapse
Affiliation(s)
- Qianyu Guo
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Jie Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Juanjuan Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Linxin Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Jia Wei
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, China
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
23
|
Hu G, Zhao X, Wang Y, Zhu X, Sun Z, Yu X, Wang J, Liu Q, Zhang J, Zhang Y, Yang J, Chang T, Ruan Z, Lv J, Gao F. Advances in B Cell Targeting for Treating Muscle-Specific Tyrosine Kinase-Associated Myasthenia Gravis. Immunotargets Ther 2024; 13:707-720. [PMID: 39678139 PMCID: PMC11646387 DOI: 10.2147/itt.s492062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024] Open
Abstract
Myasthenia gravis (MG) is a typical autoimmune disease of the nervous system. It is characterized by skeletal muscle weakness and fatigue due to impaired neuromuscular junction transmission mediated by IgG autoantibodies. Muscle-specific receptor tyrosine kinase-associated MG (MuSK-MG), a rare and severe subtype of MG, is distinguished by the presence of anti-MuSK antibodies; it responds poorly to traditional therapies. Recent research on MuSK-MG treatment has focused on specific targeted therapies. Since B cells play a critical pathogenic role in producing autoantibodies and inflammatory mediators, they are often considered the preferred target for treating MuSK-MG. Currently, various B cell-targeted drugs have been developed to treat MuSK-MG; they have shown good therapeutic effects. This review explores the evolving landscape of B cell-targeted therapies in MuSK-MG, focusing on their mechanisms, efficacy, and safety, and the current limitations associated with their use. We discuss current B cell-targeted therapies aimed at depleting or modulating B cells via both direct and indirect approaches. Furthermore, we focus on novel and promising strategies such as Chimeric Autoantibody Receptor T cell therapy, which explicitly targets MuSK-specific B cells without compromising general humoral immunity. Finally, this review provides an outlook on the potential benefits and limitations of B cell-targeted therapy in developing new therapies for MuSK-MG. We conclude by discussing future research efforts needed to optimize these therapies, expand treatment options, and improve long-term outcomes in MuSK-MG management.
Collapse
Affiliation(s)
- Guanlian Hu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- BGI College, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xue Zhao
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yiren Wang
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiaoyan Zhu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Zhan Sun
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- BGI College, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiaoxiao Yu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- BGI College, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jiahui Wang
- Department of Encephalopathy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Qian Liu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jing Zhang
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yingna Zhang
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Junhong Yang
- Department of Encephalopathy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Ting Chang
- Department of Neurology, Second Affiliated Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Zhe Ruan
- Department of Neurology, Second Affiliated Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Jie Lv
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Feng Gao
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| |
Collapse
|
24
|
Liu B, Zhao Y, Liu D, Li X, Ma Z, Yang Q. The Latest Progress in the Application of Telitacicept in Autoimmune Diseases. Drug Des Devel Ther 2024; 18:5811-5825. [PMID: 39664967 PMCID: PMC11633291 DOI: 10.2147/dddt.s493923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction Humoral immunity plays a key role in the pathogenesis of autoimmune diseases, and B-lymphocyte activating factor (BAFF) and a proliferation-inducing ligand (APRIL) are essential for the maintenance of B-lymphocyte reservoirs and humoral immunity. In March 2021, telitacicept, the world's first dual target three-channel biologic, was approved in China for the treatment of SLE and is currently in clinical trials exploring multiple indications for other autoimmune diseases. Areas Covered This article summarizes the mechanism of action, pharmacokinetics, and clinical efficacy of telitacicept for the treatment of multiple autoimmune diseases. Expert Opinion So far, the efficacy and safety of telitacicept in autoimmune diseases have been fully demonstrated in clinical practice. There are still many unresolved issues regarding the timing of initiation and discontinuation, still needs to be evaluated in future studies.
Collapse
Affiliation(s)
- Baocheng Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Yaqi Zhao
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People’s Republic of China
| | - Dongxia Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Xinya Li
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
| | - Zhenzhen Ma
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People’s Republic of China
| | - Qingrui Yang
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People’s Republic of China
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, People’s Republic of China
| |
Collapse
|
25
|
Li Z, Chen P, Zhang Y, Chen J, Zheng S, Li W, Tang L, Liu Y, Zhao N. Serum BAFF levels are associated with the prognosis of idiopathic membranous nephropathy. Ren Fail 2024; 46:2391069. [PMID: 39143819 PMCID: PMC11328817 DOI: 10.1080/0886022x.2024.2391069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVE High serum levels of B-cell activation factor (BAFF) and a proliferation-inducing ligand (APRIL) have been observed in patients with idiopathic membranous nephropathy (iMN); however, their relationships with disease severity and progression remain unclear. METHODS Patients with iMN diagnosed via renal biopsy were enrolled in this study. The concentrations of BAFF and APRIL were determined using ELISA kits. Proteinuria remission, including complete remission (CR) and partial remission (PR), and renal function deterioration were defined as clinical events. The Cox proportional hazards method was used to analyze the relationship between cytokine levels and disease progression. RESULTS Seventy iMN patients were enrolled in this study, with a median follow-up time of 24 months (range 6-72 months). The serum levels of BAFF and APRIL were higher in iMN patients than in healthy controls but lower than those in minimal change disease (MCD) patients. The serum BAFF level was positively correlated with the serum APRIL level, serum anti-phospholipase A2 receptor (anti-PLA2R) antibody level, and 24-h proteinuria and negatively correlated with the serum albumin (ALB) level. However, no significant correlation was observed between the serum APRIL level and clinical parameters. According to the multivariate Cox proportional hazards regression model adjusted for sex, age, systolic blood pressure (SBP), estimated glomerular filtration rate (eGFR), immunosuppressive agent use, 24-h proteinuria, APRIL level, and anti-PLA2R antibody, only the serum BAFF level was identified as an independent predictor of PR (HR, 0.613; 95% CI, 0.405-0.927; p = 0.021) and CR of proteinuria (HR, 0.362; 95% CI, 0.202-0.648; p < 0.001). CONCLUSIONS A high serum BAFF level is associated with severe clinical manifestations and poor disease progression in patients with iMN.
Collapse
Affiliation(s)
- Zhaohui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, P.R. China
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
| | - Ping Chen
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Ying Zhang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Juan Chen
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Shanshan Zheng
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Wenbin Li
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Lijun Tang
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Yipeng Liu
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| | - Na Zhao
- Department of Nephrology, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, P.R. China
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, P.R. China
- Shandong Institute of Nephrology, Jinan, Shandong, P.R. China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, Jinan, Shandong, P.R. China
| |
Collapse
|
26
|
Cheng X, Meng X, Chen R, Song Z, Li S, Wei S, Lv H, Zhang S, Tang H, Jiang Y, Zhang R. The molecular subtypes of autoimmune diseases. Comput Struct Biotechnol J 2024; 23:1348-1363. [PMID: 38596313 PMCID: PMC11001648 DOI: 10.1016/j.csbj.2024.03.026] [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: 11/12/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by their complexity and a wide range of clinical differences. Despite patients presenting with similar symptoms and disease patterns, their reactions to treatments may vary. The current approach of personalized medicine, which relies on molecular data, is seen as an effective method to address the variability in these diseases. This review examined the pathologic classification of ADs, such as multiple sclerosis and lupus nephritis, over time. Acknowledging the limitations inherent in pathologic classification, the focus shifted to molecular classification to achieve a deeper insight into disease heterogeneity. The study outlined the established methods and findings from the molecular classification of ADs, categorizing systemic lupus erythematosus (SLE) into four subtypes, inflammatory bowel disease (IBD) into two, rheumatoid arthritis (RA) into three, and multiple sclerosis (MS) into a single subtype. It was observed that the high inflammation subtype of IBD, the RA inflammation subtype, and the MS "inflammation & EGF" subtype share similarities. These subtypes all display a consistent pattern of inflammation that is primarily driven by the activation of the JAK-STAT pathway, with the effective drugs being those that target this signaling pathway. Additionally, by identifying markers that are uniquely associated with the various subtypes within the same disease, the study was able to describe the differences between subtypes in detail. The findings are expected to contribute to the development of personalized treatment plans for patients and establish a strong basis for tailored approaches to treating autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | - Zerun Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuai Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuhao Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hao Tang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
27
|
Saito T, Takatsuji R, Murayama G, Yamaji Y, Hagiwara Y, Nishioka Y, Kuga T, Miyashita T, Kusaoi M, Tamura N, Yamaji K. Double-filtration plasmapheresis reduces type I interferon bioavailability and inducing activity in systemic lupus erythematosus. Immunol Med 2024; 47:264-274. [PMID: 38952099 DOI: 10.1080/25785826.2024.2372918] [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: 04/22/2024] [Accepted: 06/23/2024] [Indexed: 07/03/2024] Open
Abstract
Type I interferons (IFN-Is) play a significant role in systemic lupus erythematosus (SLE) pathogenesis. Double-filtration plasmapheresis (DFPP) is a treatment option for SLE; however, its effect on IFN-Is remains unclear. Therefore, we investigated the effects of DFPP on IFN-Is. Plasma from patients with SLE (n = 11) who regularly underwent DFPP was analysed using a cell-based reporter system to detect the bioavailability and inducing activity of IFN-I. The concentration of plasma dsDNA was measured, and western blotting analysis was used to assess the phosphorylation of the STING pathway. A higher IFN-I bioavailability and inducing activity were observed in patients compared to healthy controls, and both parameters decreased after DFPP. The reduction in IFN-I-inducing activity was particularly prominent in patients with high disease activity. Notably, this reduction was not observed in STING-knockout reporter cells. Additionally, plasma dsDNA levels decreased after DFPP treatment, suggesting that inhibition of the STING pathway was responsible for the observed decrease in activity. Western blotting analysis revealed suppression of STING pathway phosphorylation after DFPP. DFPP reduced IFN-I bioavailability and the inducing activity of plasma. This reduction is likely attributable to the inhibition of the STING pathway through the elimination of dsDNA.
Collapse
Affiliation(s)
- Takumi Saito
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryo Takatsuji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Goh Murayama
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Yu Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yukitomo Hagiwara
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yujin Nishioka
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Taiga Kuga
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomoko Miyashita
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Makio Kusaoi
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
- Course of Apheresis Therapeutic Technology and Life Science, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
28
|
Hu SZ, Yuan ZY, Zhang XX, Yu XJ, Ni HY, Sun SJ, Xu T, Zhan HQ. The emerging role of BLyS/APRIL in autoimmune diseases: Biological characteristics, functions, and therapeutic potential. J Autoimmun 2024; 149:103329. [PMID: 39504927 DOI: 10.1016/j.jaut.2024.103329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024]
Abstract
Autoimmune diseases (AIDs) are common diseases in the world. Some cases are difficult to cure and can only delay the progression of the diseases. The B lymphocyte stimulator (BLyS)/a proliferation-inducing ligand (APRIL) plays an important role in B cell homeostasis, regulation of both innate and adaptive immune responses. After binding to their receptors, BLyS/APRIL primarily affects the survival and development of marginal, transitional, and mature B cells. Of note, elevated BLyS/APRIL is seen in many AIDs, such as systemic lupus erythematosus, rheumatoid arthritis, immunoglobulin A nephropathy, etc. Moreover, there is evidence that blocking these two cytokines can control the number of serum autoantibodies, promote the depletion of B lymphocytes, inhibit the activation of T cells and dendritic lymphocytes, and reduce inflammatory stress. Currently, some clinical studies are underway targeting BLyS/APRIL inhibitors for the treatment of AIDs. However, due to the scattered knowledge on the relationship between BLyS/APRIL and AIDs, it is necessary to sort out the existing data. Therefore, in this review, we describe the basic biological characteristics and functions of BLyS/APRIL in AIDs, summarize the potential clinical applications of related inhibitors, especially monoclonal antibodies and recombinant fusion proteins targeting BLyS/APRIL in AIDs, and also outline promising research directions.
Collapse
Affiliation(s)
- Shi-Zhi Hu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China; Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Zhan-Yuan Yuan
- Department of Plastic and Reconstructive Surgery, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
| | - Xiao-Xun Zhang
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Xiao-Jing Yu
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China
| | - Hai-Yan Ni
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Sheng-Jia Sun
- Clinical Medical College of Anhui Medical University, 1166 Wangjiang West Road, Hefei, Anhui, 230031, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| | - He-Qin Zhan
- Department of Pathology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui, 230601, China; Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China.
| |
Collapse
|
29
|
Croft M, Salek-Ardakani S, Ware CF. Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer. Nat Rev Drug Discov 2024; 23:939-961. [PMID: 39448880 DOI: 10.1038/s41573-024-01053-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/26/2024]
Abstract
The first anti-tumour necrosis factor (TNF) monoclonal antibody, infliximab (Remicade), celebrated its 25th anniversary of FDA approval in 2023. Inhibitors of TNF have since proved clinically efficacious at reducing inflammation associated with several autoimmune diseases, including rheumatoid arthritis, psoriasis and Crohn's disease. The success of TNF inhibitors raised unrealistic expectations for targeting other members of the TNF superfamily (TNFSF) of ligands and their receptors, with difficulties in part related to their more limited, variable expression and potential redundancy. However, there has been a resurgence of interest and investment, with many of these cytokines or their cognate receptors now under clinical investigation as targets for modulation of autoimmune and inflammatory diseases, as well as cancer. This Review assesses TNFSF-targeted biologics currently in clinical development for immune system-related diseases, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | | | - Carl F Ware
- Laboratory of Molecular Immunology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
30
|
Li S, Shi L, Zhao L, Guo Q, Li J, Liu ZL, Guo Z, Cao YJ. Split-design approach enhances the therapeutic efficacy of ligand-based CAR-T cells against multiple B-cell malignancies. Nat Commun 2024; 15:9751. [PMID: 39528513 PMCID: PMC11555413 DOI: 10.1038/s41467-024-54150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
To address immune escape, multi-specific CAR-T-cell strategies use natural ligands that specifically bind multiple receptors on malignant cells. In this context, we propose a split CAR design comprising a universal receptor expressed on T cells and ligand-based switch molecules, which preserves the natural trimeric structure of ligands like APRIL and BAFF. Following optimization of the hinges and switch labeling sites, the split-design CAR-T cells ensure the native conformation of ligands, facilitating the optimal formation of immune synapses between target cancer cells and CAR-T cells. Our CAR-T-cell strategy demonstrates antitumor activities against various B-cell malignancy models in female mice, potentially preventing immune escape following conventional CAR-T-cell therapies in the case of antigen loss or switching. This ligand-based split CAR design introduces an idea for optimizing CAR recognition, enhancing efficacy and potentially improving safety in clinical translation, and may be broadly applicable to cellular therapies based on natural receptors or ligands.
Collapse
Affiliation(s)
- Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Licai Shi
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Qiaoru Guo
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Jun Li
- Fundamenta Therapeutics Co., Ltd, Suzhou, Jiangsu, China
| | - Ze-Lin Liu
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
31
|
Su X, Yu H, Lei Q, Chen X, Tong Y, Zhang Z, Yang W, Guo Y, Lin L. Systemic lupus erythematosus: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:54. [PMID: 39472388 PMCID: PMC11522254 DOI: 10.1186/s43556-024-00217-8] [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: 07/14/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by dysregulated immune responses and autoantibody production, which affects multiple organs and varies in clinical presentation and disease severity. The development of SLE is intricate, encompassing dysregulation within the immune system, a collapse of immunological tolerance, genetic susceptibilities to the disease, and a variety of environmental factors that can act as triggers. This review provides a comprehensive discussion of the pathogenesis and treatment strategies of SLE and focuses on the progress and status of traditional and emerging treatment strategies for SLE. Traditional treatment strategies for SLE have mainly employed non-specific approaches, including cytotoxic and immunosuppressive drugs, antimalarials, glucocorticoids, and NSAIDs. These strategies are effective in mitigating the effects of the disease, but they are not a complete cure and are often accompanied by adverse reactions. Emerging targeted therapeutic drugs, on the other hand, aim to control and treat SLE by targeting B and T cells, inhibiting their activation and function, as well as the abnormal activation of the immune system. A deeper understanding of the pathogenesis of SLE and the exploration of new targeted treatment strategies are essential to advance the treatment of this complex autoimmune disease.
Collapse
Affiliation(s)
- Xu Su
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Hui Yu
- Department of Urology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400000, China
| | - Xuerui Chen
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yanli Tong
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, F-75015, France
| | - Zhongyang Zhang
- Department of Health Technology, The Danish National Research Foundation and Villum Foundation's Center IDUN, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Wenyong Yang
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Department of Neurosurgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China.
| | - Yuanbiao Guo
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Liangbin Lin
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
32
|
Richter P, Badescu MC, Rezus C, Ouatu A, Dima N, Popescu D, Burlui AM, Bratoiu I, Mihai IR, Rezus E. Antiphospholipid Antibodies as Key Players in Systemic Lupus Erythematosus: The Relationship with Cytokines and Immune Dysregulation. Int J Mol Sci 2024; 25:11281. [PMID: 39457063 PMCID: PMC11509045 DOI: 10.3390/ijms252011281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by an overproduction of cytokines, such as interleukins and interferons, contributing to systemic inflammation and tissue damage. Antiphospholipid syndrome is a thrombo-inflammatory autoimmune disease affecting a third of SLE patients. We performed an in-depth analysis of the available literature, and we highlighted the complex interplay between immunity, inflammation, and thrombosis, the three major pathogenic pathways that are trapped in a mutually reinforcing destructive loop.
Collapse
Affiliation(s)
- Patricia Richter
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
| | - Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
- IIIrd Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (A.O.); , (D.P.)
| | - Alexandra Maria Burlui
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (P.R.); (A.M.B.); (I.B.); (I.R.M.); (E.R.)
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| |
Collapse
|
33
|
Mo S, Li Y, He J, Lin L. Progress of rituximab in the treatment of systemic lupus erythematosus and lupus nephritis. Front Med (Lausanne) 2024; 11:1472019. [PMID: 39430591 PMCID: PMC11486751 DOI: 10.3389/fmed.2024.1472019] [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: 07/28/2024] [Accepted: 09/18/2024] [Indexed: 10/22/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with heterogeneous clinical manifestations, often leading to significant morbidity and mortality, particularly due to lupus nephritis (LN). The standard therapeutic approach involving mycophenolate mofetil, cyclophosphamide, and glucocorticoids has shown limitations due to cumulative toxicity and side effects. The introduction of biologic agents, especially rituximab (RTX), a chimeric monoclonal antibody targeting CD20+ B cells, has revolutionized the treatment landscape. This review synthesized the current understanding of B cells' role in SLE and LN and evaluates RTX's therapeutic impact. B cells contribute to disease pathogenesis through autoantibody production and immune complex formation, leading to tissue damage. RTX's mechanisms of action, including Complement-Dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), and induction of apoptosis, have demonstrated efficacy in both SLE and LN treatment. Clinical studies have reported remission rates and improved renal outcomes with RTX use, although challenges such as human anti-chimeric antibody development and optimal dosing persist. The review emphasized the need for continued research to elucidate RTX's long-term benefits and risks, and to explore personalized treatment strategies that incorporate B cell biology for better disease management in SLE and LN.
Collapse
Affiliation(s)
- Shouqi Mo
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Rheumatology, Jieyang People's Hospital, Jieyang, China
| | - Yilan Li
- Department of General Family Medicine, Jieyang People's Hospital, Jieyang, China
| | - Junbing He
- Jieyang Medical Research Center, Jieyang People's Hospital, Jieyang, China
| | - Ling Lin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Rheumatology, Shantou University Medical College, Shantou, China
| |
Collapse
|
34
|
Pu X, Ye Q, Zhu L, Yan T. Successful management of belimumab after obinutuzumab in a patient with systemic lupus erythematosus: a case report with an 18-month follow-up. Front Immunol 2024; 15:1459241. [PMID: 39421753 PMCID: PMC11484265 DOI: 10.3389/fimmu.2024.1459241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is a complex autoimmune disease, and despite the availability of multiple treatments, striking a balance between long-term efficacy and side effects remains a major clinical challenge. B-cell-directed therapy has attracted much attention because of its unique mechanism of action. Belimumab and obinutuzumab, as representative drugs for B-cell-directed therapy, have shown their respective advantages for SLE treatment. However, data on combination therapy with obinutuzumab and belimumab are currently unavailable. Case presentation We present the severe case report of a patient who was diagnosed with lupus nephritis (LN) with gastrointestinal involvement and developed acute renal failure. The patient responded to the first dose of obinutuzumab but failed to achieve a complete response to LN. The repeated use of obinutuzumab was limited by persistently low IgG levels and frequent infections. This is a real-world challenge that must be addressed. Therefore, the patient was subsequently treated with a novel sequential regimen of obinutuzumab followed by belimumab. After 18 months of follow-up, the patient achieved a complete clinical response with a favourable safety profile, along with the conversion of all autoantibodies from positive to negative and sustained negativity. To date, the patient has achieved a dual clinical and serological response. Conclusion There is a reason to believe that this novel combination regimen could be developed as a therapeutic strategy, with the expectation of balancing efficacy and safety.
Collapse
Affiliation(s)
- Xiuxiu Pu
- Jiaxing University Master’s Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, China
- Department of Rheumatology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qiao Ye
- Department of Rheumatology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lin Zhu
- Jiaxing University Master’s Degree Cultivation Base, Zhejiang Chinese Medical University, Jiaxing, China
- Department of Rheumatology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Tingting Yan
- Department of Rheumatology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
35
|
Moroni G, Reggiani F, Ponticelli C. Immune-mediating and immunosuppressive pharmacotherapies for proliferative lupus nephritis. Expert Opin Pharmacother 2024; 25:2061-2076. [PMID: 39402707 DOI: 10.1080/14656566.2024.2416038] [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: 08/01/2024] [Accepted: 10/09/2024] [Indexed: 11/01/2024]
Abstract
INTRODUCTION Proliferative lupus nephritis is a common and severe complication of systemic lupus erythematosus. Affected patients are at an increased risk of developing chronic kidney disease, end-stage kidney disease, and extra-renal comorbidities. In recent years, the prognosis for patients with proliferative lupus nephritis has improved thanks to advancements in management regimens. Despite these advances, lupus nephritis continues to present therapeutic complexities and unmet needs. AREAS COVERED Research was conducted across major databases to identify the most relevant articles pertaining to immune-mediating and immunosuppressive therapies in lupus nephritis. EXPERT OPINION The prognosis for patients with proliferative lupus nephritis remains severe. Some drugs used in this disease may be unable to control activity, and most of them have a low therapeutic index and may cause severe and life-threatening side effects. Nonetheless, better management of traditional drugs and the introduction of novel therapies have improved renal prognosis and reduced local and systemic adverse events in patients with proliferative lupus nephritis.
Collapse
Affiliation(s)
- Gabriella Moroni
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Francesco Reggiani
- Nephrology and Dialysis Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | |
Collapse
|
36
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
37
|
Zeng L, Yang K, Wu Y, Yu G, Yan Y, Hao M, Song T, Li Y, Chen J, Sun L. Telitacicept: A novel horizon in targeting autoimmunity and rheumatic diseases. J Autoimmun 2024; 148:103291. [PMID: 39146891 DOI: 10.1016/j.jaut.2024.103291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/19/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
BLyS and APRIL have the capability to bind to B cells within the body, allowing these cells to evade elimination when they should naturally be removed. While BLyS primarily plays a role in B cell development and maturation, APRIL is linked to B cell activation and the secretion of antibodies. Thus, in theory, inhibiting BLyS or APRIL could diminish the population of aberrant B cells that contribute to SLE and reduce disease activity in patients. Telitacicept functions by binding to and neutralizing the activities of both BLyS and APRIL, thus hindering the maturation and survival of plasma cells and fully developed B cells. The design of telitacicept is distinctive; it is not a monoclonal antibody but a TACI-Fc fusion protein generated through recombinant DNA technology. This fusion involves merging gene segments of the TACI protein, which can target BLyS/APRIL simultaneously, with the Fc gene segment of the human IgG protein. The TACI-Fc fusion protein exhibits the combined characteristics of both proteins. Currently utilized for autoimmune disease treatment, telitacicept is undergoing clinical investigations globally to assess its efficacy in managing various autoimmune conditions. This review consolidates information on the mechanistic actions, dosing regimens, pharmacokinetics, efficacy, and safety profile of telitacicept-a dual-targeted biological agent. It integrates findings from prior experiments and pharmacokinetic analyses in the treatment of RA and SLE, striving to offer a comprehensive overview of telitacicept's research advancements.
Collapse
Affiliation(s)
- Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China.
| | - Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China.
| | - Yang Wu
- Department of Rheumatology, National Clinical Research Center for Dermatologic and Immunologic Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ganpeng Yu
- People's Hospital of Ningxiang City, Ningxiang, China
| | - Yexing Yan
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Moujia Hao
- Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China
| | - Tian Song
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuwei Li
- School of Mathematics and Computational Science, Hunan University of Science and Technology, Hunan, China
| | - Junpeng Chen
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA; Psychosomatic laboratory, Department of Psychiatry, Daqing Hospital of Traditional Chinese Medicine, Daqing, China; Tong Jiecheng Studio, Hunan University of Science and Technology, Xiangtan, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Graduate School of Peking Union Medical College, Nanjing, China; Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
38
|
Qie Y, Gadd ME, Shao Q, To T, Liu A, Li S, Rivera‐Valentin R, Yassine F, Murthy HS, Dronca R, Kharfan‐Dabaja MA, Qin H, Luo Y. Targeting chronic lymphocytic leukemia with B-cell activating factor receptor CAR T cells. MedComm (Beijing) 2024; 5:e716. [PMID: 39224539 PMCID: PMC11366826 DOI: 10.1002/mco2.716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
The challenge of disease relapsed/refractory (R/R) remains a therapeutic hurdle in chimeric antigen receptor (CAR) T-cell therapy, especially for hematological diseases, with chronic lymphocytic leukemia (CLL) being particularly resistant to CD19 CAR T cells. Currently, there is no approved CAR T-cell therapy for CLL patients. In this study, we aimed to address this unmet medical need by choosing the B-cell activating factor receptor (BAFF-R) as a promising target for CAR design against CLL. BAFF-R is essential for B-cell survival and is consistently expressed on CLL tumors. Our research discovered that BAFF-R CAR T-cell therapy exerted the cytotoxic effects on both CLL cell lines and primary B cells derived from CLL patients. In addition, the CAR T cells exhibited cytotoxicity against CD19-knockout CLL cells that are resistant to CD19 CAR T therapy. Furthermore, we were able to generate BAFF-R CAR T cells from small blood samples collected from CLL patients and then demonstrated the cytotoxic effects of these patient-derived CAR T cells against autologous tumor cells. Given these promising results, BAFF-R CAR T-cell therapy has the potential to meet the long-standing need for an effective treatment on CLL patients.
Collapse
Affiliation(s)
- Yaqing Qie
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Martha E. Gadd
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Qing Shao
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Tommy To
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Andrew Liu
- Department of Cancer BiologyMayo ClinicJacksonvilleFloridaUSA
| | - Shuhua Li
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Rocio Rivera‐Valentin
- Department of Pediatric Hematology‑OncologyUniversity of Florida‐JacksonvilleJacksonvilleFloridaUSA
| | - Farah Yassine
- Division of Hematology and Medical OncologyDepartment of Internal MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Hemant S. Murthy
- Division of Hematology and Medical OncologyDepartment of Internal MedicineMayo ClinicJacksonvilleFloridaUSA
- Blood and Marrow Transplantation and Cellular Therapy ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Roxana Dronca
- Division of Hematology and Medical OncologyDepartment of Internal MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Mohamed A. Kharfan‐Dabaja
- Division of Hematology and Medical OncologyDepartment of Internal MedicineMayo ClinicJacksonvilleFloridaUSA
- Blood and Marrow Transplantation and Cellular Therapy ProgramMayo ClinicJacksonvilleFloridaUSA
| | - Hong Qin
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
- Department of Cancer BiologyMayo ClinicJacksonvilleFloridaUSA
- Division of Hematology and Medical OncologyDepartment of Internal MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Yan Luo
- Regenerative Immunotherapy and CAR‐T Translational Research ProgramMayo ClinicJacksonvilleFloridaUSA
- Department of Cancer BiologyMayo ClinicJacksonvilleFloridaUSA
| |
Collapse
|
39
|
Zhao L, Jin S, Wang S, Zhang Z, Wang X, Chen Z, Wang X, Huang S, Zhang D, Wu H. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct Target Ther 2024; 9:225. [PMID: 39198425 PMCID: PMC11358547 DOI: 10.1038/s41392-024-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Lianyu Zhao
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Song Jin
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyao Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhe Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Zhanwei Chen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Xiaohui Wang
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
40
|
Roveta A, Parodi EL, Brezzi B, Tunesi F, Zanetti V, Merlotti G, Francese A, Maconi AG, Quaglia M. Lupus Nephritis from Pathogenesis to New Therapies: An Update. Int J Mol Sci 2024; 25:8981. [PMID: 39201667 PMCID: PMC11354900 DOI: 10.3390/ijms25168981] [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: 07/11/2024] [Revised: 08/03/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Lupus Nephritis (LN) still represents one of the most severe complications of Systemic Lupus Erythematosus (SLE) and a major risk factor for morbidity and mortality. However, over the last few years, several studies have paved the way for a deeper understanding of its pathogenetic mechanisms and more targeted treatments. This review aims to provide a comprehensive update on progress on several key aspects in this setting: pathogenetic mechanisms of LN, including new insight into the role of autoantibodies, complement, vitamin D deficiency, and interaction between infiltrating immune cells and kidney resident ones; the evolving role of renal biopsy and biomarkers, which may integrate information from renal histology; newly approved drugs such as voclosporin (VOC) and belimumab (BEL), allowing a more articulate strategy for induction therapy, and other promising phase III-immunosuppressive (IS) agents in the pipeline. Several adjunctive treatments aimed at reducing cardiovascular risk and progression of chronic renal damage, such as antiproteinuric agents, represent an important complement to IS therapy. Furthermore, non-pharmacological measures concerning general lifestyle and diet should also be adopted when managing LN. Integrating these therapeutic areas requires an effort towards a holistic and multidisciplinary approach. At the same time, the availability of an increasingly wider armamentarium may translate into improvements in patient's renal outcomes over the next decades.
Collapse
Affiliation(s)
- Annalisa Roveta
- Research and Innovation Department (DAIRI), “SS Antonio e Biagio e Cesare Arrigo” University Hospital, 15121 Alessandria, Italy; (A.R.); (A.F.); (A.G.M.)
| | - Emanuele Luigi Parodi
- Nephrology and Dialysis Unit, “SS Antonio e Biagio e Cesare Arrigo” University Hospital, 15121 Alessandria, Italy; (E.L.P.); (B.B.)
| | - Brigida Brezzi
- Nephrology and Dialysis Unit, “SS Antonio e Biagio e Cesare Arrigo” University Hospital, 15121 Alessandria, Italy; (E.L.P.); (B.B.)
| | - Francesca Tunesi
- Nephrology and Dialysis Unit, IRCCS “San Raffaele” Scientific Institute, 20132 Milan, Italy;
| | - Valentina Zanetti
- Department of Internal Medicine, University of Genova, 16126 Genoa, Italy;
| | - Guido Merlotti
- Department of Primary Care, Azienda Socio Sanitaria Territoriale (ASST) of Pavia, 27100 Pavia, Italy;
| | - Alessia Francese
- Research and Innovation Department (DAIRI), “SS Antonio e Biagio e Cesare Arrigo” University Hospital, 15121 Alessandria, Italy; (A.R.); (A.F.); (A.G.M.)
| | - Antonio G. Maconi
- Research and Innovation Department (DAIRI), “SS Antonio e Biagio e Cesare Arrigo” University Hospital, 15121 Alessandria, Italy; (A.R.); (A.F.); (A.G.M.)
| | - Marco Quaglia
- Nephrology and Dialysis Unit, “SS Antonio e Biagio e Cesare Arrigo” University Hospital, 15121 Alessandria, Italy; (E.L.P.); (B.B.)
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
41
|
Chighizola CB, Suardi I, Marino A, Gattinara M, Costi S, Cattaneo A, Gerosa M, Caporali R. Belimumab-induced periungual pyogenic granulomas: A case report. Lupus 2024; 33:1017-1021. [PMID: 38860334 DOI: 10.1177/09612033241260180] [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: 06/12/2024]
Abstract
Pyogenic granuloma (PG) is a benign vascular neoformation, presenting as a painful red nodule on the skin, mucosa or nail apparatus. It is usually related to local complications such as bleedings and superinfections. The etiology of PG remains still unclear, and several triggers can lead to its formation. In case of multiple lesions, systemic conditions and drugs remain the main causes. Antineoplastic treatments, retinoids, antiretrovirals, hormones and anticonvulsants are frequently implicated in PG formation. In literature, PG has been rarely described in the course of biological treatment due to rheumatological disease. The present case report describes the development of polydactolous PGs in a 21-year-old woman with juvenile systemic lupus erythematosus (jSLE) during treatment with belimumab, a monoclonal antibody directed against BlyS. The clinical presentation, in particular the timing and the multiplicity of the lesions, and the improvement after belimumab discontinuation allowed us to consider PG as drug-induced. This case highlights the importance of considering PG as a potential complication of rheumatologic treatments.
Collapse
Affiliation(s)
- Cecilia Beatrice Chighizola
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Rheumatology and Medical Sciences, Pediatric Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Ilaria Suardi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Rheumatology and Medical Sciences, Pediatric Rheumatology Unit, ASST Pini-CTO, Milan, Italy
- Department of Rheumatology and Medical Sciences, Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Achille Marino
- Department of Rheumatology and Medical Sciences, Pediatric Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Maurizio Gattinara
- Department of Rheumatology and Medical Sciences, Pediatric Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Stefania Costi
- Department of Rheumatology and Medical Sciences, Pediatric Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Angelo Cattaneo
- Dermatology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Gerosa
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Rheumatology and Medical Sciences, Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| | - Roberto Caporali
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Department of Rheumatology and Medical Sciences, Pediatric Rheumatology Unit, ASST Pini-CTO, Milan, Italy
- Department of Rheumatology and Medical Sciences, Clinical Rheumatology Unit, ASST Pini-CTO, Milan, Italy
| |
Collapse
|
42
|
Hasegawa Y, Arinuma Y, Asakura H, Shindo R, Ino K, Kanayama Y, Tanaka T, Matsueda Y, Wada T, Oku K, Yamaoka K. Real-world efficacy of belimumab in achieving remission or low-disease activity in systemic lupus erythematosus: A retrospective study. Mod Rheumatol 2024; 34:732-740. [PMID: 37522622 DOI: 10.1093/mr/road078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/01/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
OBJECTIVES We investigated the effect of belimumab (BEL) on achieving low disease activity (LDA) and remission as an additive molecular-targeting agent to standard of care (SoC) in patients with systemic lupus erythematosus (SLE). METHODS Clinical information was retrospectively collected from patients with SLE who received BEL additive to SoC (BEL + SoC), and from patients treated with SoC alone as a control arm. Disease activity was measured by SLE-disease activity score (SLE-DAS). The proportion of patients in LDA and remission at 12 months was compared after propensity score matching. The factors contributing to LDA and remission achievement were identified by Cox proportional hazard model. RESULTS BEL + SoC significantly reduced SLE-DAS at 6 months, with a significantly higher proportion of patients achieving LDA and remission at 12 months compared to SoC alone. The presence of arthritis at baseline was significantly associated with achieving LDA and remission. Additionally, both treatment groups experienced a significant reduction in daily glucocorticoid dose. CONCLUSIONS Adding BEL to SoC was beneficial for patients with arthritis, leading to higher proportion of achieving LDA and remission, while also reducing their glucocorticoid dose. Our results indicate the utility of BEL in a treat-to-target approach for SLE patients in a real-world setting.
Collapse
Affiliation(s)
- Yasuhiro Hasegawa
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoshiyuki Arinuma
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hirotomo Asakura
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Risa Shindo
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kazuma Ino
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoshiro Kanayama
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tomoki Tanaka
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yu Matsueda
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tatsuhiko Wada
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kenji Oku
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kunihiro Yamaoka
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
43
|
Cheng J, Peng Y, Wu Q, Wu Q, He J, Yuan G. Efficacy and safety of telitacicept therapy in systemic lupus erythematosus with hematological involvement. Clin Rheumatol 2024; 43:2229-2236. [PMID: 38767710 DOI: 10.1007/s10067-024-06992-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVE To evaluate the efficacy and safety of telitacicept in SLE patients specifically with hematological involvement. METHOD A total of 22 patients with SLE and hematological involvement were included in this study. These patients received telitacicept in addition to standard therapy. We compared their demographic characteristics, clinical manifestations, and laboratory indicators before and after the administration of telitacicept. RESULTS A total of 22 patients received telitacicept treatment for a median duration of 10.4 months (ranging from 6 to 19 months). Following telitacicept therapy, significant improvements were observed in various parameters compared to baseline. Specifically, white blood cell count increased from (3.98 ± 1.80) 109/L to (6.70 ± 2.47) 109/L, (P = 0.002), hemoglobin levels increased from (100 ± 19) g/L to (125 ± 22) g/L, (P < 0.001), and platelet count increased from (83 ± 60) 109/L to (161 ± 81) 109/L, (P = 0.004). SLE Disease Activity Index (SLEDAI) scores decreased from 12(5,15) to 0(0,4), (P < 0.001). Additionally, C3 and C4 levels showed improvement. Telitacicept treatment also resulted in a significant reduction in serum IgG levels and daily prednisone dosage. Only one adverse event (4.5%) was reported during the treatment, which was a urinary tract infection. CONCLUSION The combination of telitacicept and standard treatment demonstrated significant improvements in anemia, as well as increased leukocyte and platelet levels in patients with SLE and hematological involvement. Importantly, the observed adverse events were manageable and controllable. Key Points • Telitacicept effectively improves anemia, clinical outcomes, and increases leukocyte and platelet counts. • Treatment with telitacicept leads to decreased levels of lgG, IgA, anti-dsDNA, and SLEDAI scores, while serum complement C3 and C4 returned to normal. • During the follow-up period there were observed changes in individual parameters, clinical symptoms, and organ involvement, all without significant adverse events.
Collapse
Affiliation(s)
- Jirong Cheng
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Yuanhong Peng
- Jinan University, Guangzhou, China
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Qiurong Wu
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Qian Wu
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital, 11 Xizhimen South St., Beijing, 100044, China.
| | - Guohua Yuan
- Jinan University, Guangzhou, China.
- Department of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China.
| |
Collapse
|
44
|
Wu S, Yin Y, Du L. The bidirectional relationship of depression and disturbances in B cell homeostasis: Double trouble. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110993. [PMID: 38490433 DOI: 10.1016/j.pnpbp.2024.110993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Major depressive disorder (MDD) is a recurrent, persistent, and debilitating neuropsychiatric syndrome with an increasing morbidity and mortality, representing the leading cause of disability worldwide. The dysregulation of immune systems (including innate and adaptive immune systems) has been identified as one of the key contributing factors in the progression of MDD. As the main force of the humoral immunity, B cells have an essential role in the defense against infections, antitumor immunity and autoimmune diseases. Several recent studies have suggested an intriguing connection between disturbances in B cell homeostasis and the pathogenesis of MDD, however, the B-cell-dependent mechanism of MDD remains largely unexplored compared to other immune cells. In this review, we provide an overview of how B cell abnormality regulates the progression of MMD and the potential consequence of the disruption of B cell homeostasis in patients with MDD. Abnormalities of B-cell homeostasis not only promote susceptibility to MDD, but also lead to an increased risk of developing infection, malignancy and autoimmune diseases in patients with MDD. A better understanding of the contribution of B cells underlying MDD would provide opportunities for identification of more targeted treatment approaches and might provide an overall therapeutic benefit to improve the long-term outcomes of patients with MDD.
Collapse
Affiliation(s)
- Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
45
|
Yuan C, Lin Y, Wang Y, Zhang Y, Zhao X, Yuan H, Li T, Song Q. Effects of porcine epidemic diarrhea virus infection on CD21 + B cells activation. Vet Microbiol 2024; 293:110087. [PMID: 38663176 DOI: 10.1016/j.vetmic.2024.110087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 05/15/2024]
Abstract
Porcine epidemic diarrhea virus (PEDV) is a devastating pathogen of acute- gastrointestinal infectious diseases, which can cause vomiting, diarrhea, dehydration and high morbidity and mortality among neonatal piglets. Humoral immunity plays a vital role in the host anti-PEDV infection process, but the mechanism of PEDV-induced B-cell immune response remains unknown. In this study, the effects of PEDV infection on CD21+ B cell activation were systematically analyzed through animal experiments. Enzyme-linked immunosorbent assays (ELISA) revealed that low levels of serum-specific IgA, IgM, or IgG were detected in piglets after PEDV infection, respectively. Serum interleukin (IL)-6 levels increased significantly at 4 d after infection, and the levels of IL-4, B-cell activating factor (BAFF), interferon (IFN)-γ, transforming growth factor (TGF)-β and IL-10 decreased at 7 d after infection. Fluorescence-activated cell sorting (FACS) showed that expression levels of CD21, MHC Ⅱ, CD40, and CD38 on B cell surfaces were significantly higher. In contrast, the proportions of CD21+IgM+ B cells were decreased in peripheral blood mononuclear cells (PBMCs) from the infected piglets. No differences were found in the percentage of CD21+CD80+ and CD21+CD27+ B cells in PBMCs from the infected piglets. In addition, the number of CD21+B cells in PBMCs stimulated with PEDV in vitro was significantly lower. No significant change in the mRNA expression of BCR molecules was found while the expression levels of paired immunoglobulin-like receptor B (PIR-B), B cell adaptor molecule of 32 kDa (Bam32) and BAFF were decreased. In conclusion, our research demonstrates that virulent strains of PEDV profoundly impact B cell activation, leading to alterations in phenotypic expression and BCR signaling molecules. Furthermore, this dysregulation results in compromised specific antibody secretion and perturbed cytokine production, highlighting the intricate immunological dysfunctions induced by PEDV infection.
Collapse
Affiliation(s)
- Chen Yuan
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yidan Lin
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yawen Wang
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Yanan Zhang
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Xue Zhao
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Hongxing Yuan
- Agriculture and Rural Bureau of Guantao County, Handan, Hebei Province 057750, China
| | - Tanqing Li
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China
| | - Qinye Song
- College of Veterinary Medicine, Hebei Agricultural University, China; Veterinary Biological Technology Innovation Center of Hebei Province, Baoding 071000, China.
| |
Collapse
|
46
|
Mu F, Bai X, Lou Y, Luo P, Guo Q. Rituximab alleviates pediatric systemic lupus erythematosus associated refractory immune thrombocytopenia: a case-based review. Immunol Res 2024; 72:503-511. [PMID: 38279058 DOI: 10.1007/s12026-024-09454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
A complication of pediatric systemic lupus erythematosus (pSLE) is immune thrombocytopenia (ITP). Although corticosteroids and immunoglobulins are frequently used as preliminary treatments, some patients do not respond to them. Rituximab has been reported to be safe and effective in the treatment of pSLE complicated with refractory ITP. Research is currently underway to determine the optimal rituximab dose for these individuals. We report a case of a child with SLE-associated ITP (SLE-ITP) who was successfully treated with rituximab. Rituximab is likely the most viable therapeutic option for refractory SLE-ITP. Furthermore, a comprehensive review of the relevant literature was performed and a concise overview of the pathogenesis and available treatment modalities for pediatric patients diagnosed with SLE and concurrent ITP was provided.
Collapse
Affiliation(s)
- Fangxin Mu
- Department of Nephrology and Rheumatology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Xue Bai
- Department of Nephrology and Rheumatology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yan Lou
- Department of Nephrology and Rheumatology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Ping Luo
- Department of Nephrology and Rheumatology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Qiaoyan Guo
- Department of Nephrology and Rheumatology, The Second Hospital of Jilin University, Changchun, Jilin, 130041, China.
| |
Collapse
|
47
|
Qing M, Yang D, Shang Q, Li W, Zhou Y, Xu H, Chen Q. Humoral immune disorders affect clinical outcomes of oral lichen planus. Oral Dis 2024; 30:2337-2346. [PMID: 37392455 DOI: 10.1111/odi.14667] [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: 11/01/2022] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/03/2023]
Abstract
OBJECTIVES The molecular characteristics of oral lichen planus (OLP) are still unclear, and it is not possible to distinguish the clinical outcome of OLP patients in a short period of time for follow-up. Here, we investigate the molecular characteristics of lesions in patients with stable lichen planus (SOLP) and recalcitrant erosive oral lichen planus (REOLP). METHODS Our clinical follow-up cohort was split into SOLP and REOLP groups based on the follow-up clinical data. The core modules associated with the clinical information were identified by weighted gene co-expression network analysis (WGCNA). The OLP cohort samples were divided into two groups by molecular typing, and a prediction model for OLP was created by training neural networks with the neuralnet package. RESULTS We screened 546 genes in five modules. After doing a molecular type of OLP, it was determined that B cells might have a significant impact on the clinical outcome of OLP. In addition, by means of machine learning, a prediction model was developed to predict the clinical regression of OLP with greater accuracy than the existing clinical diagnostic. CONCLUSIONS Our study revealed humoral immune disorders may make an important contribution to the clinical outcome of OLP.
Collapse
Affiliation(s)
- Maofeng Qing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Stomatology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianhui Shang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weiqi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, China
| |
Collapse
|
48
|
Pang J, Li Y, Tao R, Li J, Wang F, Xu H. Correlation Between B-Cell Activating Factor of the Tumor Necrosis Factor Family Level in Serum and Immune Inflammation in Patients with Neuropsychiatric Systemic Lupus Erythematosus and its Clinical Value. Immunol Invest 2024; 53:559-573. [PMID: 38329469 DOI: 10.1080/08820139.2024.2309567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
OBJECTIVE Neuropsychiatric systemic lupus erythematosus (NPSLE) is a form of SLE associated with severe NP syndromes causing mortality and morbidity. Respecting the fundamental of BAFF in NPSLE pathophysiology, we investigated its clinical value. METHODS Totally 105 NPSLE and 101 SLE cases without NPSLE (non-NPSLE, control) were included. Serum BAFF/TNF-α/IL-6/IL-10 levels were measured using ELISA kits. T lymphocytes were detected by flow cytometry. The independent influencing factors for NPSLE, and the auxiliary diagnostic efficacy and the ability of BAFF levels to predict adverse prognosis of NPSLE patients were analyzed by multiple factor logistic regression, and ROC curve and survival curve. RESULTS In NPSLE patients, serum BAFF level was increased and positively correlated with SLEDAI-2k, serum proinflammatory cytokines, while negatively correlated with CD4+T/CD8+T cells, and anti-inflammatory cytokine. High serum BAFF protein level was associated with a higher risk of developing NPSLE. The AUC of serum BAFF > 301.7 assisting in NPSLE diagnosis was 0.8196. Furthermore, high levels of serum BAFF were associated with a higher risk of adverse outcomes in NPSLE patients. . CONCLUSION Serum BAFF level in NPSLE patients was correlated with lymphocytes and high serum BAFF protein level could assist in diagnosis and to predict adverse outcomes in NPSLE patients.
Collapse
Affiliation(s)
- Jie Pang
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yanxia Li
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Ran Tao
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Jing Li
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Feifei Wang
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Huaheng Xu
- Department of Rheumatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
49
|
Li X, Zhao Y, Sun W, Zhang C, Yu Y, Du B, Jin A, Liu Y. Neutrophil depletion attenuates antibody-mediated rejection in a renal transplantation mouse model. Clin Exp Immunol 2024; 216:211-219. [PMID: 38150328 PMCID: PMC11036104 DOI: 10.1093/cei/uxad128] [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: 03/26/2023] [Revised: 09/05/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023] Open
Abstract
Antibody-mediated rejection (AMR) can cause graft failure following renal transplantation. Neutrophils play a key role in AMR progression, but the exact mechanism remains unclear. We investigated the effect of neutrophils on AMR in a mouse kidney transplantation model. The mice were divided into five groups: syngeneic transplantation (Syn), allograft transplantation (Allo), and three differently treated AMR groups. The AMR mouse model was established using skin grafts to pre-sensitize recipient mice. Based on the AMR model, Ly6G-specific monoclonal antibodies were administered to deplete neutrophils (NEUT-/- + AMR) and TACI-Fc was used to block B-cell-activating factor (BAFF)/a proliferation-inducing ligand (APRIL) signaling (TACI-Fc + AMR). Pathological changes were assessed using hematoxylin-eosin and immunohistochemical staining. Banff values were evaluated using the Banff 2015 criteria. Donor-specific antibody (DSA) levels were assessed using flow cytometry, and BAFF and APRIL concentrations were measured using ELISA. Compared to the Syn and Allo groups, a significantly increased number of neutrophils and increased C4d and IgG deposition were observed in AMR mice, accompanied by elevated DSA levels. Neutrophil depletion inhibited inflammatory cell infiltration and reduced C4d and IgG deposition. Neutrophil depletion significantly decreased DSA levels after transplantation and suppressed BAFF and APRIL concentrations, suggesting a mechanism for attenuating AMR-induced graft damage. Similar results were obtained after blockading BAFF/APRIL using a TACI-Fc fusion protein. In summary, neutrophil infiltration increased in the AMR mouse renal transplantation model. Neutrophil depletion or blockading the BAFF/APRIL signaling pathway significantly alleviated AMR and may provide better options for the clinical treatment of AMR.
Collapse
Affiliation(s)
- Xingku Li
- Experimental Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yakun Zhao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Wenying Sun
- Clinical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Cong Zhang
- Department of Microbiology and Immunology, College of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Yadi Yu
- Department of Immunology, College of Basic Medicine, Harbin Medical University, Harbin, People’s Republic of China
| | - Bo Du
- Experimental Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - AiShun Jin
- Department of Immunology, College of Basic Medicine, Harbin Medical University, Harbin, People’s Republic of China
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ye Liu
- Department of Immunology, College of Basic Medicine, Harbin Medical University, Harbin, People’s Republic of China
| |
Collapse
|
50
|
Yu H, Xie Y, Zuo M, Xu J, Jiang L, Liu T, Wang R, Hu D, Cha Z. Mapping theme evolution and identifying hotspots in biomarkers of systemic lupus erythematosus based on global research. Biomark Med 2024; 18:321-332. [PMID: 38648095 PMCID: PMC11218803 DOI: 10.2217/bmm-2023-0774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/12/2024] [Indexed: 04/25/2024] Open
Abstract
Objective: To perform a bibliometric analysis in the field of biomarkers for systemic lupus erythematosus. Methods: Publications were from Web of Science. Microsoft Excel, VOSviewer, Science Mapping Analysis software Tool, CiteSpace and Tableau were used for analysis. Results: A total of 1112 publications were identified; 1503 institutions from 69 countries contributed, with the highest outputs from China and Karolinska University Hospital. Petri had a tremendous impact. Academic collaborations were localized. Lupus and Arthritis & Rheumatology were the top two journals in terms of publications and citations. Lymphocyte, autoantibody, type I interferon, genetic polymorphisms and urinary biomarkers have been high-frequency themes. Conclusion: Global collaboration needs to be further strengthened. Immune cell, cytokine and gene-level research as a whole and noninvasive tests are the future trends.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Laboratory Medicine, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Yafei Xie
- West China School of Medicine/West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, 610041, China
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Meiying Zuo
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Jianguo Xu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Lili Jiang
- School of Material Science & Technology, Lanzhou University of Technology, Lanzhou, Gansu, 730050, China
| | - Ting Liu
- Department of Laboratory Medicine, Traditional Chinese Medicine Hospital of Yunyang County, Chongqing, 404500, China
| | - Renmei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Dexuan Hu
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Zhenglei Cha
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, 730000, China
| |
Collapse
|