1
|
Lee AYS, Qi Z, Jackson KJL, Reed JH. Self-reactive B cells are increased in all major stages of peripheral development in Sjögren's disease. Immunol Cell Biol 2025; 103:401-410. [PMID: 39957579 PMCID: PMC11964785 DOI: 10.1111/imcb.70005] [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: 01/14/2025] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
Sjögren's disease (SjD) is a chronic autoimmune disorder characterized by increased circulating self-reactive B cells. While many of these self-reactive B cells emerge from the bone marrow, it is not known whether they are excluded from or enriched in specific developmental stages in the periphery. The aim of this study was to determine the immunophenotype of circulating self-reactive B cells in SjD to inform more precise therapeutic targeting. Five major B cell populations: transitional, mature naïve, switched memory, double negative and plasmablasts were single-cell sorted and cultured to produce IgG. Self-reactive IgG was identified by ELISA, flow cytometry of permeabilized HEK293 cells and HEp-2 indirect immunofluorescence. Immunoglobulin heavy chains were sequenced by Sanger and next-generation sequencing. Compared with healthy donor controls (HCs), SjD patients had higher frequencies of naïve and CD21low atypical memory B cell subsets, while antigen-experienced B cells expressed more Ki67 and CD86. B cells recognizing intracellular self-antigens were identified in all stages of peripheral B cell development for SjD and HCs, but frequencies of autoreactive B cells were up to 10-fold higher in SjD. Self-reactive transitional B cells expressed higher surface CD38 and lower surface IgM. An increase in self-reactive B cells throughout peripheral development in SjD compared with HCs suggests that counterselection of autoantibody-bearing B cells during central and peripheral tolerance checkpoints are reduced in SjD. Therapeutic strategies focused on depleting B cells based on B cell receptor specificity rather than the developmental stage would be more efficient to target self-reactive B cells in SjD.
Collapse
Affiliation(s)
- Adrian YS Lee
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research and Faculty of Medicine and HealthUniversity of SydneyWestmeadNSWAustralia
- Department of ImmunologyWestmead Hospital and Institute of Clinical Pathology and Medical Research, NSW Health PathologyWestmeadNSWAustralia
| | - Zhankun Qi
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research and Faculty of Medicine and HealthUniversity of SydneyWestmeadNSWAustralia
| | | | - Joanne H Reed
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research and Faculty of Medicine and HealthUniversity of SydneyWestmeadNSWAustralia
| |
Collapse
|
2
|
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
|
3
|
Xie G, Chen X, Gao Y, Yang M, Zhou S, Lu L, Wu H, Lu Q. Age-Associated B Cells in Autoimmune Diseases: Pathogenesis and Clinical Implications. Clin Rev Allergy Immunol 2025; 68:18. [PMID: 39960645 PMCID: PMC11832777 DOI: 10.1007/s12016-025-09021-w] [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] [Accepted: 01/07/2025] [Indexed: 02/20/2025]
Abstract
As a heterogeneous B cell subset, age-associated B cells (ABCs) exhibit distinct transcription profiles, extrafollicular differentiation processes, and multiple functions in autoimmunity. TLR7 and TLR9 signals, along with IFN-γ and IL-21 stimulation, are both essential for ABC differentiation, which is also regulated by chemokine receptors including CXCR3 and CCR2 and integrins including CD11b and CD11c. Given their functions in antigen uptake and presentation, autoantibody and proinflammatory cytokine secretion, and T helper cell activation, ABCs display potential in the prognosis, diagnosis, and therapy for autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, Sjögren's syndrome, multiple sclerosis, neuromyelitis optica spectrum disorders, and ankylosing spondylitis. Specifically targeting ABCs by inhibiting T-bet and CD11c and activating CD11b and ARA2 represents potential therapeutic strategies for SLE and RA. Although single-cell sequencing technologies have recently revealed the heterogeneous characteristics of ABCs, further investigations to explore and validate ABC-target therapies are still warranted.
Collapse
Affiliation(s)
- Guangyang Xie
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Xiaojing Chen
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Yixia Gao
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Suqing Zhou
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, China.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China.
| | - Haijing Wu
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China.
- FuRong Laboratory, Changsha, China.
| | - Qianjin Lu
- Department of Dermatology, the Second Xiangya Hospital, Hunan Key Laboratory of Medical Epigenomics, Central South University, Changsha, Hunan, China.
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Chizzolini C, Guery JC, Noulet F, Gruaz L, Cenac C, Frasca L, Spoerl D, Arlettaz L, Horisberger A, Ribi C, Hugues S. Extrafollicular CD19 lowCXCR5 -CD11c - double negative 3 (DN3) B cells are significantly associated with disease activity in females with systemic lupus erythematosus. J Transl Autoimmun 2024; 9:100252. [PMID: 39444662 PMCID: PMC11497371 DOI: 10.1016/j.jtauto.2024.100252] [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: 08/21/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Objective B cells play a major role in the development and maintenance of systemic lupus erythematosus (SLE). Double negative (DN) B cells defined by the lack of surface expression of IgD and CD27 have attracted recent interest for their sensitivity to Toll-like receptor 7 (TLR7) ligands and their potential role in the production of autoantibodies. Here we aimed at investigating the possible association of DN B cells and their subsets with SLE disease activity specifically in female patients, in which TLR7 gene has been reported to escape X chromosome inactivation. Methods Peripheral blood mononuclear cells were purified from woman participating to the clinically well-characterized Swiss SLE Cohort Study (SSCS). PBMC from age-matched healthy females were used as controls. PBMC were stained for cell surface markers, intracellular Tbet and analyzed by multicolor cytofluorimetry. Single nucleotide TLR7 polymorphisms were assessed by polymerase chain reaction. Results The median SLE disease activity index of the 86 females was 2, IQR [0-6], all but 8 were under chronic SLE treatment. B cells co-expressing CD11c and Tbet were increased, the mean fluorescence intensity (MFI) of CD19 was considerably reduced and we observed a large increase in CD11c + CXCR5-and CD11c-CXCR5-concomitantly with a reduction of CD11c-CXCR5+ B cells in SLE compared to 40 healthy donors (HD). When focusing on the DN B cell subset, we found a reduction of DN1 (CD11c-CXCR5+) and an increase of DN2 (CD11c + CXCR5-) and most impressively of DN3 (CD11c-CXCR5-) cells. The DN subset, particularly DN3, showed the lowest level of CD19 expression. Both DN1 and DN3 percentages as well as the CD19 MFI of DN cells were associated with SLE disease activity. The use of glucocorticoids, immunosuppressants, and antimalarials impacted differentially on the frequencies of DN B cell subsets. CD19 MFI in B cells and the percentage of DN3 were the strongest biomarkers of disease activity. The TLR7 snp3858384 G allele was associated with increased percentages of B cells and CD19+CD11c-CXCR5+ and decreased CD19+CD11c-CXCR5-. Conclusions DN3 B cells are strongly associated with SLE clinical activity pointing to their potential involvement in disease pathogenesis, and CD19 expression level performs accurately as disease activity biomarker.
Collapse
Affiliation(s)
- Carlo Chizzolini
- Department of Pathology and Immunology, Centre Médical Universitaire, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Jean-Charles Guery
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291, CNRS UMR5051, University Paul Sabatier Toulouse, F-31024, Toulouse, France
| | - Fanny Noulet
- Department of Pathology and Immunology, Centre Médical Universitaire, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Lyssia Gruaz
- Department of Pathology and Immunology, Centre Médical Universitaire, School of Medicine, University of Geneva, Geneva, Switzerland
| | - Claire Cenac
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity) INSERM UMR1291, CNRS UMR5051, University Paul Sabatier Toulouse, F-31024, Toulouse, France
| | - Loredana Frasca
- National Center for Global Health, Istituto Superiore di Sanità, Rome, Italy
| | - David Spoerl
- Clinical Immunology and Allergy, Department of Medicine, University Hospital and Faculty of Medicine, Geneva, Switzerland
| | - Lionel Arlettaz
- Department of Biology, ICH, Valais Hospital, Sion, Switzerland
| | - Alice Horisberger
- Service of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Camillo Ribi
- Service of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Stéphanie Hugues
- Department of Pathology and Immunology, Centre Médical Universitaire, School of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
5
|
Reyes-Huerta RF, Mandujano-López V, Velásquez-Ortiz MG, Alcalá-Carmona B, Ostos-Prado MJ, Reyna-Juárez Y, Meza-Sánchez DE, Juárez-Vega G, Mejía-Domínguez NR, Torres-Ruiz J, Gómez-Martín D, Maravillas-Montero JL. Novel B-cell subsets as potential biomarkers in idiopathic inflammatory myopathies: insights into disease pathogenesis and disease activity. J Leukoc Biol 2024; 116:84-94. [PMID: 38554062 DOI: 10.1093/jleuko/qiae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
Idiopathic inflammatory myopathies are a heterogeneous group of rare autoimmune disorders characterized by progressive muscle weakness and the histopathologic findings of inflammatory infiltrates in muscle tissue. Although their pathogenesis remains indefinite, the association of autoantibodies with clinical manifestations and the evidence of high effectiveness of depleting therapies suggest that B cells could be implicated. Therefore, we explored the landscape of peripheral B cells in this disease by multiparametric flow cytometry, finding significant numerical decreases in memory and double-negative subsets, as well as an expansion of the naive compartment relative to healthy controls, that contribute to defining disease-associated B-cell subset signatures and correlating with different clinical features of patients. Additionally, we determined the potential value of these subsets as diagnostic biomarkers, thus positioning B cells as neglected key elements possibly participating in idiopathic inflammatory myopathy onset or development.
Collapse
Affiliation(s)
- Raúl F Reyes-Huerta
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, CU, Coyoacán, Mexico City 04510, Mexico
| | - Vladimir Mandujano-López
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
| | - Ma Guadalupe Velásquez-Ortiz
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Unidad de Posgrado, CU, Coyoacán, Mexico City 04510, Mexico
| | - Beatriz Alcalá-Carmona
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - María J Ostos-Prado
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Yatzil Reyna-Juárez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - David E Meza-Sánchez
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Jiram Torres-Ruiz
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - Diana Gómez-Martín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| | - José L Maravillas-Montero
- B cell Immunology Laboratory, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Circuito de la Investigación Científica S/N, CU, Coyoacán, Mexico City 04510, Mexico
- Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán y Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
6
|
Gartshteyn Y, Geraldino-Pardilla L, Khalili L, Bukhari S, Lerrer S, Winchester RJ, Askanase AD, Mor A. SAP-expressing T peripheral helper cells identify systemic lupus erythematosus patients with lupus nephritis. Front Immunol 2024; 15:1327437. [PMID: 38550577 PMCID: PMC10972949 DOI: 10.3389/fimmu.2024.1327437] [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: 10/24/2023] [Accepted: 02/21/2024] [Indexed: 04/02/2024] Open
Abstract
Introduction T follicular (TFH) and peripheral helper (TPH) cells have been increasingly recognized as a pathogenic subset of CD4 T cells in systemic lupus erythematosus (SLE). The SLAM Associated Protein (SAP) regulates TFH and TPH function by binding to the co-stimulatory signaling lymphocyte activation molecule family (SLAMF) receptors that mediate T cell - B cell interactions. SAP and SLAMF are critical for TPH-dependent B cell maturation into autoantibody-producing plasma cells that characterize SLE pathogenesis. We hypothesized that SAP-expressing TPH cells are involved in the pathogenesis of lupus nephritis (LN). Methods Peripheral blood mononuclear cells (PBMC) were isolated using density gradient separation from whole blood. Cells were stained for cell surface markers, followed by permeabilization and staining of intracellular SAP for spectral flow cytometry analysis. We also analyzed SAP expression from renal infiltrating LN T cells using the available single-cell RNA sequencing (scRNA seq) Accelerated Medicines Partnership (AMP) SLE dataset. Results PBMC from 30 patients with SLE (34 ± 10 years old, 83% female), including 10 patients with LN, were analyzed. We found an increase in total SAP-positive CD4 and CD8 T cells in SLE compared with controls (55.5 ± 2.6 vs. 41.3 ± 3.4, p=0.007, and 52.5 ± 3.0 vs. 39.2 ± 2.8, p=0.007 respectively). In CD4 T cells, the highest SAP expression was in the TPH subset. The frequency of SAP+TPH in circulation correlated with disease activity; SLE patients with renal disease had higher levels of circulating SAP+TPH that remained significant after adjusting for age, sex, race, low complements, and elevated anti-dsDNA (p=0.014). scRNA-seq data of renal infiltrating T cells in LN identified SAP expression to localize to the TFH-like CD4 cluster and GZMK+ CD8 cluster. Increased SAP expression in LN was associated with the differential expression of SLAMF3 and SLAMF7 and granzyme K and EOMES. The existence of two predominant SAP-expressing subsets, the TFH-like CD4 T cells, and GZMK+ effector CD8 T cells, was verified using scRNA-seq data from a human transcriptomic atlas of fifteen major organs. Conclusion The expansion of SAP-expressing T helper cells was associated with LN in our cohort and verified using scRNA-seq data of renal infiltrating T cells. Improved SLAM and SAP signaling understanding can identify new therapeutic targets in LN.
Collapse
Affiliation(s)
- Yevgeniya Gartshteyn
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Laura Geraldino-Pardilla
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Leila Khalili
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Shoiab Bukhari
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Shalom Lerrer
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| | - Robert J. Winchester
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Anca D. Askanase
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Adam Mor
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
7
|
He Y, Vinuesa CG. Germinal center versus extrafollicular responses in systemic autoimmunity: Who turns the blade on self? Adv Immunol 2024; 162:109-133. [PMID: 38866437 PMCID: PMC7616122 DOI: 10.1016/bs.ai.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Spontaneously formed germinal centers (GCs) have been reported in most mouse models of human autoimmune disease and autoimmune patients, and have long been considered a source of somatically-mutated and thus high affinity autoantibodies, but their role in autoimmunity is becoming increasingly controversial, particularly in the context of systemic autoimmune diseases like lupus. On the one hand, there is good evidence that some pathogenic lupus antibodies have acquired somatic mutations that increase affinity for self-antigens. On the other hand, recent studies that have genetically prevented GC formation, suggest that GCs are dispensable for systemic autoimmunity, pointing instead to pathogenic extrafollicular (EF) B-cell responses. Furthermore, several lines of evidence suggest germinal centers may in fact be somewhat protective in the context of autoimmunity. Here we review how some of the conflicting evidence arose, and current views on the role of GCs in autoimmunity, outlining mechanisms by which GC may eliminate self-reactivity. We also discuss recent advances in understanding extrafollicular B cell subsets that participate in autoimmunity.
Collapse
Affiliation(s)
- Yuke He
- China-Australia Centre for Personalised Immunology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Carola G Vinuesa
- China-Australia Centre for Personalised Immunology, Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China; Francis Crick Institute, London, United Kingdom.
| |
Collapse
|
8
|
Tang YY, Wang DC, Chen YY, Xu WD, Huang AF. Th1-related transcription factors and cytokines in systemic lupus erythematosus. Front Immunol 2023; 14:1305590. [PMID: 38164134 PMCID: PMC10757975 DOI: 10.3389/fimmu.2023.1305590] [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: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory disorder related to immunity dysfunction. The Th1 cell family including Th1 cells, transcription factor T-bet, and related cytokines IFNγ, TNFα, IL-2, IL-18, TGF-β, and IL-12 have been widely discussed in autoimmunity, such as SLE. In this review, we will comprehensively discuss the expression profile of the Th1 cell family in both SLE patients and animal models and clarify how the family members are involved in lupus development. Interestingly, T-bet-related age-associated B cells (ABCs) and low-dose IL-2 treatment in lupus were emergently discussed as well. Collection of the evidence will better understand the roles of the Th1 cell family in lupus pathogenesis, especially targeting IL-2 in lupus.
Collapse
Affiliation(s)
- Yang-Yang Tang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Da-Cheng Wang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - You-Yue Chen
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
9
|
Vinuesa CG, Shen N, Ware T. Genetics of SLE: mechanistic insights from monogenic disease and disease-associated variants. Nat Rev Nephrol 2023; 19:558-572. [PMID: 37438615 DOI: 10.1038/s41581-023-00732-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 07/14/2023]
Abstract
The past few years have provided important insights into the genetic architecture of systemic autoimmunity through aggregation of findings from genome-wide association studies (GWAS) and whole-exome or whole-genome sequencing studies. In the prototypic systemic autoimmune disease systemic lupus erythematosus (SLE), monogenic disease accounts for a small fraction of cases but has been instrumental in the elucidation of disease mechanisms. Defects in the clearance or digestion of extracellular or intracellular DNA or RNA lead to increased sensing of nucleic acids, which can break B cell tolerance and induce the production of type I interferons leading to tissue damage. Current data suggest that multiple GWAS SLE risk alleles act in concert with rare functional variants to promote SLE development. Moreover, introduction of orthologous variant alleles into mice has revealed that pathogenic X-linked dominant and recessive SLE can be caused by novel variants in TLR7 and SAT1, respectively. Such bespoke models of disease help to unravel pathogenic pathways and can be used to test targeted therapies. Cell type-specific expression data revealed that most GWAS SLE risk genes are highly expressed in age-associated B cells (ABCs), which supports the view that ABCs produce lupus autoantibodies and contribute to end-organ damage by persisting in inflamed tissues, including the kidneys. ABCs have thus emerged as key targets of promising precision therapeutics.
Collapse
Affiliation(s)
- Carola G Vinuesa
- The Francis Crick Institute, London, UK.
- University College London, London, UK.
- China Australia Centre for Personalized Immunology (CACPI), Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China.
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
- Center for Autoimmune Genomics and Aetiology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Paediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Thuvaraka Ware
- The Francis Crick Institute, London, UK
- University College London, London, UK
| |
Collapse
|
10
|
李 永, 吴 小. [Research progress in systemic lupus erythematosus from 2021 to 2022]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:785-790. [PMID: 37668024 PMCID: PMC10484082 DOI: 10.7499/j.issn.1008-8830.2302150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/03/2023] [Indexed: 09/06/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects multiple organ systems, presenting a complex and diverse clinical manifestation. The heterogeneous treatment response and prognosis of SLE pose significant challenges to its diagnosis, classification, and homogeneous treatment. The emergence of new technologies and fields, such as synthetic biology, genomics, and proteomics, has contributed to a deeper exploration of the pathogenesis and biomarkers of SLE, facilitating precision diagnosis and treatment. This review summarizes the latest research data and achievements in SLE for the years 2021-2022, providing an overview and summary of relevant studies conducted in the past two years.
Collapse
|
11
|
Moysidou E, Lioulios G, Christodoulou M, Xochelli A, Stai S, Iosifidou M, Iosifidou A, Briza S, Briza DI, Fylaktou A, Stangou M. Increase in Double Negative B Lymphocytes in Patients with Systemic Lupus Erythematosus in Remission and Their Correlation with Early Differentiated T Lymphocyte Subpopulations. Curr Issues Mol Biol 2023; 45:6667-6681. [PMID: 37623240 PMCID: PMC10453294 DOI: 10.3390/cimb45080421] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/26/2023] Open
Abstract
B and T lymphocytes demonstrate important alterations in patients with systemic lupus erythematous (SLE), with a significant upregulation of double negative (DN) B cells. The aim of this study was to evaluate the correlation of T cell immunity changes with the distinct B-cell-pattern SLE. In the present study, flow cytometry was performed in 30 patients in remission of SLE and 31 healthy controls to detect DN B cells (CD19+IgD-CD27-) and a wide range of T lymphocyte subpopulations based on the presence of CD45RA, CCR7, CD31, CD28, and CD57, defined as naive, memory, and advanced differentiated/senescent T cells. Both B and T lymphocytes were significantly reduced in SLE patients. However, the percentage of DN B cells were increased compared to HC (12.9 (2.3-74.2) vs. 8 (1.7-35), p = 0.04). The distribution of CD4 and CD8 lymphocytes demonstrated a shift to advanced differentiated subsets. The population of DN B cells had a significant positive correlation with most of the early differentiated T lymphocytes, CD4CD31+, CD4CD45RA+CD28+, CD4CD45RA+CD57-, CD4CD45RA-CD57-, CD4CD28+CD57-, CD4CD28+CD57+, CD4 CM, CD8 CD31+, CD8 NAÏVE, CD8CD45RA-CD57-, CD8CD28+CD57-, and CD8CD28+CD57+. Multiple regression analysis revealed CD4CD31+, CD8CD45RA-CD57-, and CD8CD28+CD57- cells as independent parameters contributing to DN B cells, with adjusted R2 = 0.534 and p < 0.0001. The predominance of DN B cells in patients with SLE is closely associated with early differentiated T lymphocyte subsets, indicating a potential causality role of DN B cells in T lymphocyte activation.
Collapse
Affiliation(s)
- Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
- Department of Nephrology, General Hospital “Hippokration”, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
- Department of Nephrology, General Hospital “Hippokration”, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
- Department of Nephrology, General Hospital “Hippokration”, 54642 Thessaloniki, Greece
| | - Aliki Xochelli
- Department of Immunology, National Histocompatibility Center, General Hospital “Hippokration”, 54642 Thessaloniki, Greece; (A.X.); (A.F.)
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
- Department of Nephrology, General Hospital “Hippokration”, 54642 Thessaloniki, Greece
| | - Myrto Iosifidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
| | - Artemis Iosifidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
| | - Sophia Briza
- Department of Architecture, School of Engineering, University of Thessaly, 38334 Thessaly, Greece;
| | - Dimitria Ioanna Briza
- School of Informatics, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece;
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, General Hospital “Hippokration”, 54642 Thessaloniki, Greece; (A.X.); (A.F.)
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thessaloniki, Greece; (E.M.); (G.L.); (M.C.); (S.S.); (M.I.); (A.I.)
- Department of Nephrology, General Hospital “Hippokration”, 54642 Thessaloniki, Greece
| |
Collapse
|
12
|
Qu L, Jiao B. The Interplay between Immune and Metabolic Pathways in Kidney Disease. Cells 2023; 12:1584. [PMID: 37371054 PMCID: PMC10296595 DOI: 10.3390/cells12121584] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Kidney disease is a significant health problem worldwide, affecting an estimated 10% of the global population. Kidney disease encompasses a diverse group of disorders that vary in their underlying pathophysiology, clinical presentation, and outcomes. These disorders include acute kidney injury (AKI), chronic kidney disease (CKD), glomerulonephritis, nephrotic syndrome, polycystic kidney disease, diabetic kidney disease, and many others. Despite their distinct etiologies, these disorders share a common feature of immune system dysregulation and metabolic disturbances. The immune system and metabolic pathways are intimately connected and interact to modulate the pathogenesis of kidney diseases. The dysregulation of immune responses in kidney diseases includes a complex interplay between various immune cell types, including resident and infiltrating immune cells, cytokines, chemokines, and complement factors. These immune factors can trigger and perpetuate kidney inflammation, causing renal tissue injury and progressive fibrosis. In addition, metabolic pathways play critical roles in the pathogenesis of kidney diseases, including glucose and lipid metabolism, oxidative stress, mitochondrial dysfunction, and altered nutrient sensing. Dysregulation of these metabolic pathways contributes to the progression of kidney disease by inducing renal tubular injury, apoptosis, and fibrosis. Recent studies have provided insights into the intricate interplay between immune and metabolic pathways in kidney diseases, revealing novel therapeutic targets for the prevention and treatment of kidney diseases. Potential therapeutic strategies include modulating immune responses through targeting key immune factors or inhibiting pro-inflammatory signaling pathways, improving mitochondrial function, and targeting nutrient-sensing pathways, such as mTOR, AMPK, and SIRT1. This review highlights the importance of the interplay between immune and metabolic pathways in kidney diseases and the potential therapeutic implications of targeting these pathways.
Collapse
Affiliation(s)
- Lili Qu
- Division of Nephrology, Department of Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030-1405, USA
| | - Baihai Jiao
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030-1405, USA
| |
Collapse
|
13
|
Perl A, Agmon-Levin N, Crispín JC, Jorgensen TN. Editorial: New biomarkers for the diagnosis and treatment of systemic lupus erythematosus. Front Immunol 2022; 13:1009038. [PMID: 36311710 PMCID: PMC9599399 DOI: 10.3389/fimmu.2022.1009038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/20/2022] [Indexed: 01/17/2023] Open
Affiliation(s)
- Andras Perl
- Department of Medicine, College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, United States,Department of Biochemistry and Molecular Biology, College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, United States,Department of Microbiology and Immunology, College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY, United States,*Correspondence: Andras Perl,
| | - Nancy Agmon-Levin
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Ramat Gan, Israel
| | - José C. Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico,Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Mexico
| | - Trine N. Jorgensen
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|