1
|
Sowerby JM, Rao DA. T cell-B cell interactions in human autoimmune diseases. Curr Opin Immunol 2025; 93:102539. [PMID: 40020254 PMCID: PMC11927756 DOI: 10.1016/j.coi.2025.102539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/11/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
Activation of autoreactive B cells and production of specific autoantibodies are hallmark features of many autoimmune diseases. B cell differentiation into antibody-secreting cells typically requires help from cognate T cells, which provide both cytokines and cell surface signals in an intricate intercellular interaction. A range of T cells can provide this help to B cells, including T follicular helper cells in follicles of secondary lymphoid organs, as well as T peripheral helper cells, which accumulate within inflamed target tissues in autoimmune diseases. Here, we discuss recent observations about the phenotypes of B cell-helper T cells that accumulate in inflamed tissues and in circulation of patients with autoimmune diseases, the correlations between B cell-helper T cells and B cells in these tissues, and key mediators of productive T cell-B cell interactions, with a focus on mediators that are being targeted therapeutically. Understanding the scope of B cell-helper T cells and their functions will improve our ability to quantify and track pathologic T cell-B cell interactions in human autoimmune diseases and may highlight critical mediators that can be targeted to suppress these interactions therapeutically.
Collapse
Affiliation(s)
- John M Sowerby
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, USA.
| |
Collapse
|
2
|
Wang X, Wen B, Duan X, Zhang Y, Hu Y, Li H, Shang H, Jing Y. Recent Advances of Type I Interferon on the Regulation of Immune Cells and the Treatment of Systemic Lupus Erythematosus. J Inflamm Res 2025; 18:4533-4549. [PMID: 40182060 PMCID: PMC11967359 DOI: 10.2147/jir.s516195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease with multiple organ damage. Several studies have found that, in addition to significant production of autoantibodies, the majority of SLE patients exhibit increased expression of type I interferon (IFN-I) regulated genes (also known as IFN-I traits), and that IFN-I plays a crucial role in the pathogenesis of SLE. In SLE, virtually all immune cells are dysregulated, and most of these aberrant dysregulations are directly or indirectly affected by IFN-I. The mechanism of action of IFN-I in these immune cells is multifaceted. In this review, we focus on the immune cell types that produce IFN-I and are affected by IFN-I in SLE. Importantly, we explore the research progress of related drugs in terms of IFN-I production, itself, and downstream. Here we provide the most up-to-date information on the mechanisms that lead to the pathogenesis of SLE, providing the basis for the development of innovative future therapies and future research directions.
Collapse
Affiliation(s)
- Xiaocui Wang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Bin Wen
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People’s Republic of China
| | - Xuemei Duan
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Yunfei Zhang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Ying Hu
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Haonan Li
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| | - Huifeng Shang
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, People’s Republic of China
| | - Yukai Jing
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, People’s Republic of China
| |
Collapse
|
3
|
Yu H, Ma Z, Su S, Xu Z, Yi H. RNA modification: a promising code to unravel the puzzle of autoimmune diseases and CD4 + T cell differentiation. Front Immunol 2025; 16:1563150. [PMID: 40196109 PMCID: PMC11973318 DOI: 10.3389/fimmu.2025.1563150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Dynamic changes in various forms of RNA modification are critical to the functional homeostasis of the immune system and the pathophysiology of autoimmune diseases. RNA modification-related proteins play an essential role in these processes. At present, the research methods of RNA modification in autoimmune diseases are mainly to detect the expression changes of RNA modification-related proteins in tissues or cells, but there is a lack of explorations of target RNAs and in-depth mechanisms. Considering the important role of CD4+ T cell dysfunction in the pathogenesis and progression of autoimmune diseases, the regulatory effect of abnormal RNA modification on CD4+ T cells deserves attention, which will provide a perspective for further exploring the mechanism of RNA modification in autoimmune diseases. In this Review, we discuss the abnormal RNA modification changes in patients with autoimmune diseases and highlight the effects of these abnormal changes on CD4+ T cells.
Collapse
Affiliation(s)
- Hui Yu
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| | - Zhanchuan Ma
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| | - Sensen Su
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| | - Zheng Xu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huanfa Yi
- Central Laboratory, The First Hospital of Jilin University, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| |
Collapse
|
4
|
Dai X, Fan Y, Zhao X. Systemic lupus erythematosus: updated insights on the pathogenesis, diagnosis, prevention and therapeutics. Signal Transduct Target Ther 2025; 10:102. [PMID: 40097390 PMCID: PMC11914703 DOI: 10.1038/s41392-025-02168-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/26/2024] [Accepted: 01/26/2025] [Indexed: 03/19/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory illness with heterogeneous clinical manifestations covering multiple organs. Diversified types of medications have been shown effective for alleviating SLE syndromes, ranging from cytokines, antibodies, hormones, molecular inhibitors or antagonists, to cell transfusion. Drugs developed for treating other diseases may benefit SLE patients, and agents established as SLE therapeutics may be SLE-inductive. Complexities regarding SLE therapeutics render it essential and urgent to identify the mechanisms-of-action and pivotal signaling axis driving SLE pathogenesis, and to establish innovative SLE-targeting approaches with desirable therapeutic outcome and safety. After introducing the research history of SLE and its epidemiology, we categorized primary determinants driving SLE pathogenesis by their mechanisms; combed through current knowledge on SLE diagnosis and grouped them by disease onset, activity and comorbidity; introduced the genetic, epigenetic, hormonal and environmental factors predisposing SLE; and comprehensively categorized preventive strategies and available SLE therapeutics according to their functioning mechanisms. In summary, we proposed three mechanisms with determinant roles on SLE initiation and progression, i.e., attenuating the immune system, restoring the cytokine microenvironment homeostasis, and rescuing the impaired debris clearance machinery; and provided updated insights on current understandings of SLE regarding its pathogenesis, diagnosis, prevention and therapeutics, which may open an innovative avenue in the fields of SLE management.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China.
| | - Yuting Fan
- Tissue Engineering and Stem Cell Experiment Center, Tumor Immunotherapy Technology Engineering Research Center, Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, P. R. China
- Department of Gastroenterology, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, P. R. China
| | - Xing Zhao
- Tissue Engineering and Stem Cell Experiment Center, Tumor Immunotherapy Technology Engineering Research Center, Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, P. R. China.
| |
Collapse
|
5
|
Du Y, Faz-Lopez B, Ah Kioon MD, Cenac C, Pierides M, Lakin KS, Spiera RF, Chaumeil J, Truchetet ME, Gordon JK, Guéry JC, Barrat FJ. Altered X-chromosome inactivation of the TLR7/8 locus and heterogeneity of pDCs in systemic sclerosis. J Exp Med 2025; 222:e20231809. [PMID: 39670995 PMCID: PMC11639950 DOI: 10.1084/jem.20231809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that has a strong female predominance. Both the X-linked TLR7 and TLR8 can induce type I IFN (IFN-I) by plasmacytoid DCs (pDCs), which can promote fibrosis. We identified five subclusters of pDCs, including ISGhigh clusters that were over-represented in SSc patients. We observed that both TLR7 and TLR8 genes escape from X chromosome inactivation (XCI) at higher frequency in pDCs of SSc patients, which was associated with changes in TLR7 protein profile. Combined DNA/RNA FISH analysis revealed that the TLR7/8 locus is preferentially located outside of the inactive X (Xi) territory when TLR7 is expressed, suggesting that higher-order loop formation is linked to TLR7/8 expression from the Xi. Furthermore, the expression levels of XIST and the transcriptional repressor SPEN were reduced in SSc pDCs. Hence, our data revealed the heterogeneity of pDCs in SSc and suggested that altered XCI at the TLR7/8 locus may contribute to the chronic IFN-I activity of pDCs in female SSc patients.
Collapse
Affiliation(s)
- Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Bérénice Faz-Lopez
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Claire Cenac
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Michael Pierides
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Kimberly S. Lakin
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Robert F. Spiera
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Julie Chaumeil
- Institut Cochin, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Marie-Elise Truchetet
- ImmunoConcEpt, CNRS, UMR 5164, University of Bordeaux, Talence, France
- Rheumatology Department, CHU de Bordeaux, Bordeaux, France
| | - Jessica K. Gordon
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Jean-Charles Guéry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
6
|
Attrill MH, Shinko D, Viveiros TM, Milighetti M, de Gruijter NM, Jebson B, Kartawinata M, Rosser EC, Wedderburn LR, Pesenacker AM. Treg fitness signatures as a biomarker for disease activity in Juvenile Idiopathic Arthritis. J Autoimmun 2025; 152:103379. [PMID: 39954509 DOI: 10.1016/j.jaut.2025.103379] [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: 10/16/2024] [Revised: 01/06/2025] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
Juvenile Idiopathic Arthritis (JIA) is an autoimmune condition characterised by flares of joint inflammation. However, no reliable biomarker exists to predict the erratic disease course. Normally, regulatory T cells (Tregs) maintain tolerance, with altered Tregs associated with autoimmunity. Treg signatures have shown promise in monitoring other conditions, therefore a Treg gene/protein signature could offer novel biomarker potential for predicting disease activity in JIA. Machine learning on our nanoString Treg 48-gene signature on peripheral blood (PB) Tregs generated a model to distinguish active JIA (active joint count, AJC≥1) Tregs from healthy controls (HC, AUC = 0.9875 on test data). Biomarker scores from this model successfully differentiated inactive (AJC = 0) from active JIA PB Tregs. Moreover, scores correlated with clinical activity scores (cJADAS), and discriminated subclinical disease (AJC = 0, cJADAS≥0.5) from remission (cJADAS<0.5). To investigate altered protein expression as a surrogate measure for Treg fitness in JIA, we utilised spectral flow cytometry and unbiased clustering analysis. Three Treg clusters were of interest in active JIA PB, including TIGIThighCD226highCD25low Teff-like Tregs, CD39-TNFR2-Helioshigh, and a 4-1BBlowTIGITlowID2intermediate Treg cluster predominated in inactive JIA PB (AJC = 0). The ratio of these Treg clusters correlated to cJADAS, and higher ratios could potentially predict inactive individuals that flared by 9-month follow-up. Thus, we demonstrate altered Treg signatures and subsets as an important factor, and useful biomarker, for disease progression versus remission in JIA, revealing genes and proteins contributing to Treg fitness. Ultimately, PB Treg fitness measures could serve as routine biomarkers to guide disease and treatment management to sustain remission in JIA.
Collapse
Affiliation(s)
- Meryl H Attrill
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK; Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK
| | - Diana Shinko
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK
| | - Telma Martins Viveiros
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK
| | - Martina Milighetti
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK; Cancer Institute, UCL, London, WC1E 6DD, UK
| | - Nina M de Gruijter
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; Division of Medicine, UCL, London, WC1E 6JF, UK
| | - Bethany Jebson
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK
| | - Melissa Kartawinata
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK
| | - Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; Division of Medicine, UCL, London, WC1E 6JF, UK
| | - Lucy R Wedderburn
- Infection, Immunity & Inflammation Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, UCL, London, WC1N 1EH, UK; Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, London, WC1E 6JF, UK; NIHR Biomedical Research Centre at GOSH, London, WC1N 1EH, UK
| | - Anne M Pesenacker
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, London, NW3 2PP, UK.
| |
Collapse
|
7
|
Kume M, Din J, Zegarra-Ruiz DF. Dysregulated Intestinal Host-Microbe Interactions in Systemic Lupus Erythematosus: Insights from Patients and Mouse Models. Microorganisms 2025; 13:556. [PMID: 40142449 PMCID: PMC11944652 DOI: 10.3390/microorganisms13030556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by chronic inflammation that affects multiple organs, with its prevalence varying by ethnicity. Intestinal dysbiosis has been observed in both SLE patients and murine models. Additionally, intestinal barrier impairment is thought to contribute to the ability of pathobionts to evade and breach immune defenses, resulting in antigen cross-reactivity, microbial translocation, subsequent immune activation, and, ultimately, multiple organ failure. Since the detailed mechanisms underlying these processes are difficult to examine using human samples, murine models are crucial. Various SLE murine models, including genetically modified spontaneous and inducible murine models, offer insights into pathobionts and how they dysregulate systemic immune systems. Furthermore, since microbial metabolites modulate systemic immune responses, bacteria and their metabolites can be targeted for treatment. Based on human and mouse research insights, this review examines how lupus pathobionts trigger intestinal and systemic immune dysregulation. Therapeutic approaches, such as fecal microbiota transplantation and dietary adjustments, show potential as cost-effective and safe methods for preventing and treating SLE. Understanding the complex interactions between the microbiota, host factors, and immune dysregulation is essential for developing novel, personalized therapies to tackle this multifaceted disease.
Collapse
Affiliation(s)
| | | | - Daniel F. Zegarra-Ruiz
- Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA; (M.K.); (J.D.)
| |
Collapse
|
8
|
Kyttaris V, Wallace DJ, Khosroshahi A, Concoff A, Wilson N, Liu CC, Manzi S, Ahearn J, Taghavi S, Warsi T, Park S, Schleif C, Partain BD, O’Malley T. Multi-centered clinical validation demonstrating superior precision in lupus diagnosis: T cell autoantibodies and TC4d outperform conventional lupus erythematosus biomarkers. Front Immunol 2025; 16:1518208. [PMID: 40093011 PMCID: PMC11907206 DOI: 10.3389/fimmu.2025.1518208] [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: 10/28/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction T Cell autoantibodies, TIgG and TIgM, as well as the T Cell-bound complement protein fragment C4d (TC4d) are novel diagnostic biomarkers that have demonstrated high specificity and sensitivity for SLE. The present study aims to characterize the clinical performance characteristics of the emergent T Cell biomarkers in a multi-center clinical validation cohort. Methods A cohort of 400 adult patients enrolled across 3 academic and 2 community-based autoimmune rheumatic centers, comprised of 105 SLE patients, 173 patients with autoimmune rheumatic diseases (ARD), 83 apparently healthy volunteers (AHV) and 39 other (non-autoimmune) disease (OD) controls were tested for TC4d, TIgG, TIgM and an extensive autoantibody profile. Diagnostic specificity was assessed against the ARD, AHV and OD groups, individually. Semi-quantitative flow cytometry analysis included TIgG and TIgM autoantibodies, cell-bound complement activation products (CB-CAPs), TC4d, erythrocyte-bound C4d (EC4d) and B lymphocyte-bound C4d (BC4d). Conventional autoantibodies and soluble complement proteins, C3 and C4, were assessed by ELISA and immunoturbidimetry, respectively. Results ROC analysis distinguishing ANA-positive (ANA+) SLE (N = 91) from ARD, TIgG, BC4d and TC4d demonstrated AUC values 0.81, 0.80 and 0.79, respectively, outperforming anti-dsDNA (0.72), C3 (0.69), TIgM (0.67), C4 (0.66) and anti-Smith (0.61). A similar ranking of discriminatory power was observed in ROC analysis distinguishing ANA+ SLE vs. OD as well as ANA+ SLE vs. AHV. At 95% diagnostic specificity for SLE vs. AHV, the sensitivity (95% CI) of TC4d, TIgG and TIgM for SLE was 58.1% (48.1 - 67.7%), 31.4% (22.7 - 41.2%) and 29.5% (21.0 - 39.2%), respectively. The T Cell SLE biomarkers uniquely identified 19% (20/105) of SLE patients who were otherwise negative (serologically inactive) for conventional SLE autoantibodies and had normal serum complement levels. Among the serologically inactive SLE subset, the T Cell SLE biomarkers collectively identified 53% of subjects. Conclusions The novel SLE biomarkers TC4d, TIgG and TIgM consistently outperform conventional markers across multiple cohorts. Their integration enhances diagnostic sensitivity, especially in SLE-specific autoantibody negative patients with normal complement levels. When coupled with conventional biomarkers, these novel tests may enable earlier and more accurate SLE detection, leading to more timely diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | | | - Nicole Wilson
- Allegheny Health Network, Pittsburgh, PA, United States
| | | | - Susan Manzi
- Allegheny Health Network, Pittsburgh, PA, United States
| | - Joseph Ahearn
- Allegheny Health Network, Pittsburgh, PA, United States
| | | | | | | | | | | | | |
Collapse
|
9
|
Georgakis S, Ioannidou K, Mora BB, Orfanakis M, Brenna C, Muller YD, Del Rio Estrada PM, Sharma AA, Pantaleo G, de Leval L, Comte D, Gottardo R, Petrovas C. Cellular and molecular determinants mediating the dysregulated germinal center immune dynamics in systemic lupus erythematosus. Front Immunol 2025; 16:1530327. [PMID: 40070830 PMCID: PMC11894538 DOI: 10.3389/fimmu.2025.1530327] [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: 11/18/2024] [Accepted: 01/17/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is characterized by dysregulated humoral immunity, leading to the generation of autoreactive B cells that can differentiate both within and outside of lymph node (LN) follicles. Methods Here, we employed spatial transcriptomics and multiplex imaging to investigate the follicular immune landscaping and the in situ transcriptomic profile in LNs from SLE individuals. Results Our spatial transcriptomic analysis revealed robust type I IFN and plasma cell signatures in SLE compared to reactive, control follicles. Cell deconvolution revealed that follicular T cell subsets are mainly affected by the type I IFN fingerprint of SLE follicles. Dysregulation of TFH differentiation was documented by i) the significant reduction of Bcl6hi TFH cells, ii) the reduced cell density of potential IL-4 producing TFH cell subsets associated with the impaired transcriptomic signature of follicular IL-4 signaling and iii) the loss of their correlation with GC-B cells. This profile was accompanied by a marked reduction of Bcl6hi B cells and an enrichment of extrafollicular CD19hiCD11chiTbethi, age-associated B cells (ABCs), known for their autoreactive potential. The increased prevalence of follicular IL-21hi cells further reveals a hyperactive microenvironment in SLE compared to control. Discussion Taken together, our findings highlight the altered immunological landscape of SLE follicles, likely fueled by potent inflammatory signals such as sustained type I IFN and/or IL-21 signaling. Our work provides novel insights into the spatial molecular and cellular signatures of SLE follicular B and TFH cell dynamics, and points to druggable targets to restore immune tolerance and enhance vaccine responses in SLE patients.
Collapse
Affiliation(s)
- Spiros Georgakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Kalliopi Ioannidou
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Bernat Bramon Mora
- Biomedical Data Science Center, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Cloe Brenna
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Yannick D. Muller
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Perla M. Del Rio Estrada
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Ashish A. Sharma
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Denis Comte
- Service of Internal Medicine, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raphael Gottardo
- Biomedical Data Science Center, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
- Swiss Institute for Bioinformatics, Lausanne, Switzerland
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| |
Collapse
|
10
|
Liu X, Zhang Y, Wang Y, Xu Y, Xia W, Liu R, Xu S. Inflammatory Gene Signature Identified by Machine Algorithms Reveals Novel Biomarkers of Coronary Artery Disease. J Inflamm Res 2025; 18:2033-2044. [PMID: 39959641 PMCID: PMC11827506 DOI: 10.2147/jir.s496046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/30/2025] [Indexed: 02/18/2025] Open
Abstract
Purpose Inflammatory activation of immune cells plays a pivotal role in the development of coronary artery diseases (CAD). This study aims to investigate the immune responses of peripheral blood mononuclear cells (PBMCs) in CAD and to identify novel signature genes and biomarkers using machine learning algorithms. Methods The GSE113079 dataset was analyzed and differentially expressed genes (DEGs) were identified between CAD and normal samples. The intersection of DEGs with inflammation-related genes was used to identify the differentially expressed inflammation-related genes (DIRGs). Then, the receiver operating characteristic (ROC) curves were plotted for each DIRG, and those with an area under the curve (AUC) greater than 0.9 were selected for subsequent analysis. Furthermore, machine learning algorithms were employed to identify biomarkers. A nomogram was developed based on these biomarkers. The CIBERSORT algorithm and Wilcoxon test method were used to analyze the differences in immune cells between the CAD and normal samples. The identified biomarkers were validated in PBMCs from patients with CAD and in atherosclerotic aortas from ApoE-/- mice. Results A total of 574 DEGs were identified between CAD and normal samples. From this intersection, 29 DIRGs were identified, of which 14 DIRGs (PTGER1, IL17RC, KLKB1, GPR32, ADM, NUPR1, SCN9A, IL17B, CX3CL1, FFAR3, PYDC2, SYT11, RORA, and GPR31) exhibited high diagnostic efficacy. Four biomarkers (ADM, NUPR1, PTGER1, and PYDC2) were identified using Support Vector Machine (SVM). Ten types of immune cells, including CD8+ T cells, regulatory T cells (Tregs), and resting NK cells, showed significant differences between the CAD and normal groups. Furthermore, increased levels of ADM, NUPR1, PTGER1, and PYDC2 were validated in PBMCs isolated from CAD patients. In addition, ADM, NUPR1, and PTGER1 were upregulated in the mouse atherosclerotic aorta. Conclusion Our findings revealed novel inflammatory gene signatures of CAD that could be potential biomarkers for the early diagnosis of CAD.
Collapse
Affiliation(s)
- Xing Liu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuanyuan Zhang
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Yan Wang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- Health Management Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Yanfeng Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- National - Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Wenhao Xia
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- National - Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- Department of Cardiovascular Medicine, Guangxi Hospital Division of The First Affiliated Hospital of Sun Yat-sen University, Nanning, Guangxi, People’s Republic of China
| | - Ruiming Liu
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- National - Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Shiyue Xu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- National - Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
11
|
Cardon A, Guinebretière T, Dong C, Gil L, Ado S, Gavlovsky PJ, Braud M, Danger R, Schultheiß C, Doméné A, Paul-Gilloteaux P, Chevalier C, Bernier L, Judor JP, Fourgeux C, Imbert A, Khaldi M, Bardou-Jacquet E, Elkrief L, Lannes A, Silvain C, Schnee M, Tanne F, Vavasseur F, Brusselle L, Brouard S, Kwok WW, Mosnier JF, Lohse AW, Poschmann J, Binder M, Gournay J, Conchon S, Milpied P, Renand A. Single cell profiling of circulating autoreactive CD4 T cells from patients with autoimmune liver diseases suggests tissue imprinting. Nat Commun 2025; 16:1161. [PMID: 39880819 PMCID: PMC11779892 DOI: 10.1038/s41467-025-56363-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Autoimmune liver diseases (AILD) involve dysregulated CD4 T cell responses against liver self-antigens, but how these autoreactive T cells relate to liver tissue pathology remains unclear. Here we perform single-cell transcriptomic and T cell receptor analyses of circulating, self-antigen-specific CD4 T cells from patients with AILD and identify a subset of liver-autoreactive CD4 T cells with a distinct B-helper transcriptional profile characterized by PD-1, TIGIT and HLA-DR expression. These cells share clonal relationships with expanded intrahepatic T cells and exhibit transcriptional signatures overlapping with tissue-resident T cells in chronically inflamed environments. Using a mouse model, we demonstrate that, following antigen recognition in the liver, CD4 T cells acquire an exhausted phenotype, play a crucial role in liver damage, and are controlled by immune checkpoint pathways. Our findings thus suggest that circulating autoreactive CD4 T cells in AILD are imprinted by chronic antigen exposure to promote liver inflammation, thereby serving as a potential target for developing biomarkers and therapies for AILD.
Collapse
Affiliation(s)
- Anaïs Cardon
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Thomas Guinebretière
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Chuang Dong
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, CIML, Marseille, France
| | - Laurine Gil
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, CIML, Marseille, France
| | - Sakina Ado
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, CIML, Marseille, France
| | - Pierre-Jean Gavlovsky
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Martin Braud
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Richard Danger
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Christoph Schultheiß
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Aurélie Doméné
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | | | | | - Laura Bernier
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Jean-Paul Judor
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Cynthia Fourgeux
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Astrid Imbert
- Service Hepato-gastro-entérologie et Assistance Nutritionnelle, CHU Nantes, Nantes, France
| | - Marion Khaldi
- Service Hepato-gastro-entérologie et Assistance Nutritionnelle, CHU Nantes, Nantes, France
- Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Nantes, France
| | - Edouard Bardou-Jacquet
- CHU Rennes, Service des maladies du foie, Université Rennes, INSERM, INRAE, Institut NUMECAN, Rennes, France
| | - Laure Elkrief
- CHRU Tours, Service Hépato-Gastroentérologie, Tours, France
| | - Adrien Lannes
- CHU Angers, Service Hépato-Gastroentérologie et Oncologie Digestive, Université d'Angers, Laboratoire HIFIH, UPRES EA3859, SFR 4208, Angers, France
| | | | - Matthieu Schnee
- CHD Vendée-La Roche sur Yon, Service Hépato-Gastroentérologie, F- 85000, la Roche sur Yon, France
| | - Florence Tanne
- CHU Brest, Service Hépato-Gastroentérologie, Brest, France
| | | | - Lucas Brusselle
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Sophie Brouard
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Jean-François Mosnier
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- Service Anatomie et Cytologie Pathologiques, CHU Nantes, Nantes, France
| | - Ansgar W Lohse
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jeremie Poschmann
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Mascha Binder
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Jérôme Gournay
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- Service Hepato-gastro-entérologie et Assistance Nutritionnelle, CHU Nantes, Nantes, France
- Institut des Maladies de l'Appareil Digestif, IMAD, CHU Nantes, Nantes, France
| | - Sophie Conchon
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
| | - Pierre Milpied
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy, CIML, Marseille, France.
| | - Amédée Renand
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
| |
Collapse
|
12
|
Sasaki T, Sowerby J, Xiao Y, Wang R, Marks KE, Horisberger A, Gao Y, Lee PY, Qu Y, Sze MA, Alves SE, Levesque MC, Fujio K, Costenbader KH, Rao DA. Clonal relationships between Tph and Tfh cells in patients with SLE and in murine lupus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635189. [PMID: 39974998 PMCID: PMC11838332 DOI: 10.1101/2025.01.27.635189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Pathologic T cell-B cell interactions drive disease in systemic lupus erythematosus (SLE). The T cells that activate B cell responses include T peripheral helper (Tph) and T follicular helper (Tfh) cells, yet the developmental and clonal relationships between these B cell-helper T cell populations are unclear. Here we use T cell receptor (TCR) profiling to demonstrate clonal overlap between Tph and Tfh cells in the circulation of patients with SLE. Expanded Tph and Tfh cell clones persist over the course of 1 year in patients with a new diagnosis of SLE, and clones are observed to shift both from Tfh to Tph cells and from Tph to Tfh cells over time. High resolution analysis of cells sorted as Tph cells (CXCR5- PD-1hi) and Tfh cells (CXCR5+ PD-1hi) from SLE patients revealed considerable heterogeneity among cells sorted as Tph cells and highlighted a specific cluster of cells that expressed transcriptomic features of activated B cell-helper T cells. This cell population, marked by expression of TOX and CXCL13, was found in both sorted Tph and Tfh cells, and was clonally linked in these two populations. Analysis of B cell-helper T cells in murine pristane-induced lupus demonstrated similar populations of Tph and Tfh cells in both lung and spleen with strong clonal overlap. T cell-specific loss of Bcl6 prevented accumulation of Tfh cells and reduced accumulation of Tph cells in pristane-treated mice, indicating a role for Bcl6 in the survival and expansion of both populations. Together, these observations demonstrate a shared developmental path among pathologically expanded Tph and Tfh cells in lupus. The persistence of expanded Tph and Tfh cells clones over time may impose barriers to induction of stable tolerance by immunosuppressive medications or by B cell depletion.
Collapse
Affiliation(s)
- Takanori Sasaki
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - John Sowerby
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yinan Xiao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Runci Wang
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathryne E Marks
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alice Horisberger
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yidan Gao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yujie Qu
- Merck & Co., Inc., Boston, MA, USA
| | | | | | | | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Faliti CE, Van TTP, Anam FA, Cheedarla N, Williams ME, Mishra AK, Usman SY, Woodruff MC, Kraker G, Runnstrom MC, Kyu S, Sanz D, Ahmed H, Ghimire M, Morrison-Porter A, Quehl H, Haddad NS, Chen W, Cheedarla S, Neish AS, Roback JD, Antia R, Hom J, Tipton CM, Lindner JM, Ghosn E, Khurana S, Scharer CD, Khosroshahi A, Lee FEH, Sanz I. Disease-associated B cells and immune endotypes shape adaptive immune responses to SARS-CoV-2 mRNA vaccination in human SLE. Nat Immunol 2025; 26:131-145. [PMID: 39533072 PMCID: PMC11695260 DOI: 10.1038/s41590-024-02010-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 mRNA vaccination has reduced effectiveness in certain immunocompromised individuals. However, the cellular mechanisms underlying these defects, as well as the contribution of disease-induced cellular abnormalities, remain largely unexplored. In this study, we conducted a comprehensive serological and cellular analysis of patients with autoimmune systemic lupus erythematosus (SLE) who received the Wuhan-Hu-1 monovalent mRNA coronavirus disease 2019 vaccine. Our findings revealed that patients with SLE exhibited reduced avidity of anti-receptor-binding domain antibodies, leading to decreased neutralization potency and breadth. We also observed a sustained anti-spike response in IgD-CD27- 'double-negative (DN)' DN2/DN3 B cell populations persisting during memory responses and with greater representation in the SLE cohort. Additionally, patients with SLE displayed compromised anti-spike T cell immunity. Notably, low vaccine efficacy strongly correlated with higher values of a newly developed extrafollicular B and T cell score, supporting the importance of distinct B cell endotypes. Finally, we found that anti-BAFF blockade through belimumab treatment was associated with poor vaccine immunogenicity due to inhibition of naive B cell priming and an unexpected impact on circulating T follicular helper cells.
Collapse
Affiliation(s)
- Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Trinh T P Van
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Fabliha A Anam
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Narayanaiah Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - M Elliott Williams
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ashish Kumar Mishra
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD, USA
| | - Sabeena Y Usman
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | | | - Martin C Runnstrom
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Shuya Kyu
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Daniel Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Hasan Ahmed
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Midushi Ghimire
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Andrea Morrison-Porter
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Hannah Quehl
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Natalie S Haddad
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
- MicroB-plex, Inc., Atlanta, GA, USA
| | - Weirong Chen
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Suneethamma Cheedarla
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - John D Roback
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Jennifer Hom
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Christopher M Tipton
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | | | - Eliver Ghosn
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration, Silver Spring, MD, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Arezou Khosroshahi
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| |
Collapse
|
14
|
McDermott MM, Walter KL, Bibbins-Domingo K. Translational Science Reviews-A New JAMA Review. JAMA 2024; 332:1888. [PMID: 39495501 DOI: 10.1001/jama.2024.21146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Affiliation(s)
- Mary M McDermott
- Division of General Internal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | |
Collapse
|
15
|
Dörner T, Lipsky PE. The essential roles of memory B cells in the pathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol 2024; 20:770-782. [PMID: 39511302 DOI: 10.1038/s41584-024-01179-5] [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: 10/02/2024] [Indexed: 11/15/2024]
Abstract
Emerging evidence indicates that memory B cells are dysfunctional in systemic lupus erythematosus (SLE). They are hyporesponsive to signalling through the B cell receptor (BCR) but retain responsiveness to Toll-like receptor (TLR) and type I interferon signalling, as well as to T cell-mediated activation via CD40-CD154. Chronic exposure to immune complexes of ribonucleoprotein (RNP)-specific autoantibodies and TLR-engaging or BCR-engaging cargo is likely to contribute to this partially anergic phenotype. TLR7 or TLR8 signalling and the resulting production of type I interferon, as well as the sustained activation by bystander T cells, fuel a positive feedforward loop in memory B cells that can evade negative selection and permit preferential expansion of anti-RNP autoantibodies. Clinical trials of autologous stem cell transplantation or of B cell-targeted monoclonal antibodies and chimeric antigen receptor (CAR) T cells have correlated replenishment of the memory B cell population with relapse of SLE. Moreover, the BCR hyporesponsiveness of memory B cells might explain the failure of non-depleting B cell-targeting approaches in SLE, including BTK inhibitors and anti-CD22 monoclonal antibodies. Thus, targeting of dysfunctional memory B cells might prove effective in SLE, while also avoiding the adverse events of broad-spectrum targeting of B cell and plasma cell subsets that are not directly involved in disease pathogenesis.
Collapse
Affiliation(s)
- Thomas Dörner
- Department Medicine/Rheumatology and Clinical Immunology, Charite Universitätsmedizin Berlin & Deutsches Rheumaforschungszentrum (DRFZ), Berlin, Germany.
| | | |
Collapse
|
16
|
Nettelfield S, Chen Z. Unraveling the origins of pathogenic CXCL13 + helper T cells in systemic lupus erythematosus. Immunol Cell Biol 2024; 102:757-759. [PMID: 39218440 DOI: 10.1111/imcb.12818] [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: 09/04/2024]
Abstract
This Research Highlight discusses a recent publication, where the authors identified an increase in CXCL13+ peripheral helper T/follicular helper T cells, which was concomitant with a decrease in CD96+ T helper 22 (TH22) cells in patients with systemic lupus erythematosus. The genetic and epigenetic cues that reciprocally regulate this pathogenic imbalance of T-cell subsets were also identified, thus providing targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sam Nettelfield
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Zhian Chen
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
17
|
Choi MY, Costenbader KH, Fritzler MJ. Environment and systemic autoimmune rheumatic diseases: an overview and future directions. Front Immunol 2024; 15:1456145. [PMID: 39318630 PMCID: PMC11419994 DOI: 10.3389/fimmu.2024.1456145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Despite progress in our understanding of disease pathogenesis for systemic autoimmune rheumatic diseases (SARD), these diseases are still associated with high morbidity, disability, and mortality. Much of the strongest evidence to date implicating environmental factors in the development of autoimmunity has been based on well-established, large, longitudinal prospective cohort studies. Methods Herein, we review the current state of knowledge on known environmental factors associated with the development of SARD and potential areas for future research. Results The risk attributable to any particular environmental factor ranges from 10-200%, but exposures are likely synergistic in altering the immune system in a complex interplay of epigenetics, hormonal factors, and the microbiome leading to systemic inflammation and eventual organ damage. To reduce or forestall the progression of autoimmunity, a better understanding of disease pathogenesis is still needed. Conclusion Owing to the complexity and multifactorial nature of autoimmune disease, machine learning, a type of artificial intelligence, is increasingly utilized as an approach to analyzing large datasets. Future studies that identify patients who are at high risk of developing autoimmune diseases for prevention trials are needed.
Collapse
Affiliation(s)
- May Y Choi
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
| | - Karen H Costenbader
- Department of Medicine, Div of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, United States
- Medicine, Harvard Medical School, Boston, MA, United States
| | - Marvin J Fritzler
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
18
|
Pazhouhandeh M, Yu D. Interferon disrupts immune and tissue homeostasis in SLE via CXCL13. Nat Rev Rheumatol 2024:10.1038/s41584-024-01164-y. [PMID: 39251769 DOI: 10.1038/s41584-024-01164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Affiliation(s)
- Mehrdad Pazhouhandeh
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Di Yu
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
19
|
Cen K, Zhou J, Yang X, Guo Y, Xiao Y. Lymphocyte antigen 6 family member E suppresses apoptosis and promotes pancreatic cancer growth and migration via Wnt/β-catenin pathway activation. Sci Rep 2024; 14:20196. [PMID: 39215036 PMCID: PMC11364638 DOI: 10.1038/s41598-024-70764-1] [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: 06/10/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Pancreatic cancer (PC) is the primary cause of cancer-related mortality. Due to the absence of reliable biomarkers for predicting prognosis or guiding treatment, there is an urgent need for molecular studies on PC. Lymphocyte antigen 6 family member E (LY6E) is implicated in uncontrolled cell growth across various cancers. However, the precise mechanism of LY6E in PC remains unclear. Here, we conducted comprehensive bioinformatic analyses using online tools and R- × 64-4.1.1, complemented by experimental validation through Western blotting, immunohistochemistry, immunosorbent assays, flow cytometry, cell assays, and animal models. Our findings reveal significantly elevated expression of LY6E in PC, correlating with poor prognosis. LY6E knockdown inhibited proliferation, invasion, and migration of PC cells, while enhancing apoptosis evidenced by increased cleaved caspase 3 levels and alterations in the Bcl-2/Bax ratio. Conversely, LY6E overexpression promoted PC cell proliferation and migration, and inhibited apoptosis. Mechanistically, LY6E downregulation suppressed the Wnt/β-catenin signaling pathway. In vivo studies demonstrated that LY6E suppression attenuated tumor growth in murine models. Additionally, LY6E suppression resulted in reduced tumor growth in mice. In conclusion, our study confirms the significant role of LY6E in the progression of PC. LY6E, serving as an independent prognostic indicator, has the potential to serve as a valuable biomarker for PC to inform treatment strategies.
Collapse
Affiliation(s)
- Kenan Cen
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jingyao Zhou
- Department of Pharmacy, Taizhou Central Hospital, Taizhou, China
| | - Xuejia Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yangyang Guo
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yanyi Xiao
- The Dingli Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Shanghai University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|