1
|
McGrath S, Sundbeck B, Thorarinsdottir K, Jonsson CA, Camponeschi A, Agelii ML, Ekwall AKH, Hetland ML, Østergaard M, Uhlig T, Nurmohamed M, Lampa J, Nordström D, Hørslev-Petersen K, Gudbjornsson B, Gröndal G, van Vollenhoven R, Rudin A, Mårtensson IL, Gjertsson I. Transitional and CD21 - PD-1 + B cells are associated with remission in early rheumatoid arthritis. BMC Rheumatol 2025; 9:45. [PMID: 40259340 PMCID: PMC12010607 DOI: 10.1186/s41927-025-00487-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Early initiation of effective treatment is associated with positive long-term prognosis for patients with rheumatoid arthritis (RA). Currently, there are no biomarkers in clinical use to predict treatment response. A predictor of treatment response may be the B-cell compartment, as this is altered in RA patients, making it a potential candidate for predicting treatment response. In this study, we sought to identify B-cell subset(s) at diagnosis that might be associated with Clinical Disease Activity Index (CDAI) remission at 24-week follow-up. METHODS Seventy early RA patients from the NORD-STAR trial, recruited from two Swedish sites, and 28 matched healthy controls, were included in this spin-off study. In NORD-STAR, all patients were randomized to methotrexate (MTX) combined with 1) prednisolone, 2) anti-TNF (certolizumab-pegol), 3) CTLA4-Ig (abatacept), or 4) anti-IL-6R (tocilizumab). Circulating B-cell subsets at diagnosis were assessed by flow cytometry. The primary outcome measure was remission according to CDAI ≤ 2.8. A multivariate two-part discriminant analysis was performed to assess whether B-cell subpopulations at diagnosis could predict remission at 24 weeks. Subsequent univariable statistical analyses were performed using t-tests, Mann-Whitney U, or Kruskal-Wallis tests, as appropriate. Correlations were analyzed using Spearman or Pearson tests, depending on data type. The impact of specific B-cell populations on remission at week 24 was assessed using logistic regression models. The logistic regression model was also used to simultaneously visualize the sensitivity and specificity of the model for all possible values of the exposure (B-cell subpopulations) in predicting the outcome. RESULTS Patients who achieved CDAI remission at 24 weeks had higher proportions of transitional (p < 0.01) and CD21- PD-1+ (p < 0.01) B cells at diagnosis compared to those who did not. When the two B-cell populations were combined, the sensitivity and specificity for remission, including all treatment arms, were 59% and 86%, respectively. Stratification of the patients by treatment arm revealed a significant negative correlation between the proportion of transitional B cells at baseline and disease activity after 24 weeks of treatment with either MTX and prednisolone or anti-IL-6R. CONCLUSIONS Our results indicate that transitional and CD21- PD-1+ B cells are associated with remission in early RA. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Sarah McGrath
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Boel Sundbeck
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden.
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Katrin Thorarinsdottir
- Center for Rheumatology Research, Department of Rheumatology, Landspitali University Hospital, Reykjavik, Iceland
| | - Charlotte A Jonsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Alessandro Camponeschi
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Monica Leu Agelii
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin H Ekwall
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Merete Lund Hetland
- Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Østergaard
- Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Centre for Head and Orthopaedics, Rigshospitalet, Glostrup, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Till Uhlig
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway
- University of Oslo, Oslo, Norway
| | - Michael Nurmohamed
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Rheumatology and Immunology Center, Reade, Amsterdam, The Netherlands
| | - Jon Lampa
- Division of Rheumatology, Department of Medicine, Center for Molecular Medicine (CMM), Karolinska Institute, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
| | - Dan Nordström
- Division of Medicine and Rheumatology, Helsinki University Hospital, Helsinki, Finland
| | - Kim Hørslev-Petersen
- Danish Hospital for Rheumatic Diseases, University Hospital of Southern Denmark, Sønderborg, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Bjorn Gudbjornsson
- Center for Rheumatology Research, Department of Rheumatology, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Gerdur Gröndal
- Center for Rheumatology Research, Department of Rheumatology, Landspitali University Hospital, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ronald van Vollenhoven
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Anna Rudin
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Inga-Lill Mårtensson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Gothenburg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
2
|
Fang G, Chen J, Xi T, Liu Y, Wu Y, Wen Y, Tang H. A cohort of highly activated CD99 - CD72 + B cells promoting autoimmune progression in juvenile systemic lupus erythematosus. Int Immunopharmacol 2025; 152:114466. [PMID: 40090085 DOI: 10.1016/j.intimp.2025.114466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/17/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025]
Abstract
CD72 inhibits the development of systemic lupus erythematosus (SLE) by suppressing TLR7-dependent B cell responses to self-nucleic acids (NAs). The absence of CD72 promotes the progression of lupus disease. Here, we find a highly activated subset of CD99- CD72+ B cells (CD72+ BCs) expressing elevated levels of TLR7 in juvenile SLE, which contributes to autoimmunity. Through multi-omics integrated analysis of single-cell RNA sequencing(scRNA-seq) data and bulk RNA sequencing (RNA-seq) data, we demonstrate that CD72+ BCs possess characteristics of both activated naïve B cells (aN) and age-associated B cells (ABCs). Concurrently, CD72+ BCs exhibit pronounced plasmablast-like features compared to other cellular subpopulations. We propose a plausible conclusion that CD72+ BCs represent a critical transitional cell population involved in the activation and subsequent plasma cell differentiation of naïve B cells and age-related B cells following exposure to self-antigens in SLE. This finding offers novel opportunities for elucidating the genesis of autoantibody-secreting cells involved in the autoimmune response processes in lupus.
Collapse
Affiliation(s)
- Guofeng Fang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science Technology, Wuhan 430030, China
| | - Jing Chen
- Department of Rheumatology and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Ting Xi
- Department of Rheumatology and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Yi Liu
- Department of Rheumatology and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Yali Wu
- Department of Rheumatology and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Yini Wen
- Department of Rheumatology and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China
| | - Hongxia Tang
- Department of Rheumatology and Immunology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430016, China.
| |
Collapse
|
3
|
Li X, Zhu Q, Yang Z, Zhu M, Xie Z, Fan Y, Zhao T. The Jieduquyuziyin prescription alleviates systemic lupus erythematosus by modulating B cell metabolic reprogramming via the AMPK/PKM2 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119626. [PMID: 40081513 DOI: 10.1016/j.jep.2025.119626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Jieduquyuziyin prescription (JP) is an enhanced formula derived from the "Sheng Ma Bie Jia Tang" in the Golden Chamber. JP is an empirical formula approved for use in the treatment of systemic lupus erythematosus (SLE) in hospitals across China, demonstrating notable therapeutic effects. It has been shown to suppress B cell activation and alleviate symptoms; however, its underlying mechanism remains unclear. AIM OF THE STUDY The aim of this study is to investigate the effect of JP on B cell metabolic reprogramming in the treatment of SLE and to elucidate the underlying regulatory mechanisms. MATERIALS AND METHODS Core targets and pathways regulating B cell activation were identified through sequencing of activated and resting B cells from SLE patients and network pharmacology of JP. Targeted metabolomics was employed to assess JP's effect on B cell metabolism. In vivo experiments evaluated the effects of JP, JP combined with a PKM2 inhibitor, or an AMPK inhibitor on SLE activity, B cell activation and glycolysis. RESULTS Bioinformatics analyses identified PKM as a core target in B cell activation, which was significantly upregulated and linked to the inhibition of AMPK signaling. JP reduced B cell glycolysis and activation, leading to significant improvements in disease pathology. The combination of JP with an AMPK inhibitor diminished the therapeutic effect. Further studies suggested that JP inhibits glycolysis-dependent B cell activation via the AMPK/PKM2 pathway, reducing germinal center responses and effector B cells. CONCLUSION This study reveals that the AMPK/PKM2 pathway is a promising therapeutic target for regulating immune metabolic imbalance in SLE B cells. Additionally, it provides evidence that JP may improve SLE by activating the AMPK/PKM2 pathway to inhibit glycolysis-dependent B cell activation, laying the foundation for further investigation of its therapeutic mechanisms.
Collapse
Affiliation(s)
- Xiaolong Li
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qingmiao Zhu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Zi Yang
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Mengyu Zhu
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - ZhiJun Xie
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, 310053, China
| | - Yongsheng Fan
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 311500, China
| | - Ting Zhao
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Research Institute of Chinese Medical Clinical Foundation and Immunology, Hangzhou, 310053, China.
| |
Collapse
|
4
|
Ziyadullaev SK, Khudaiberdiev SS, Aripova TU, Chirumbolo S, Kamalov ZS, Bjørklund G, Rizaev JA, Tashkenbaeva EN, Khamidov OA, Gaffarov UB. Synovial Fluid as a Crucial Component of the Joint Microenvironment in Rheumatoid Arthritis. Immune Netw 2025; 25:e2. [PMID: 40342839 PMCID: PMC12056296 DOI: 10.4110/in.2025.25.e2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/17/2024] [Accepted: 12/22/2024] [Indexed: 05/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease closely associated with synovial tissue proliferation, pannus formation in small joints such as the hands, wrists, and feet, cartilage destruction, and systemic complications such as pulmonary, cardiovascular, neurological, and skeletal muscle lesions, glucocorticoid-induced osteoporosis and infections. The importance of confirming the diagnosis and determining local activity is given to the study of synovial fluid. A deep understanding of the pathological process in the joint in RA, characterized by changes in autoreactive CD4+ T cells, B cells, macrophages, inflammatory cytokines, chemokines, and autoantibodies, has now been achieved, although much remains to be explored. This article provides an updated overview of the pathogenesis of RA, revealing even more therapeutic targets for the intra-articular pathological process.
Collapse
Affiliation(s)
| | | | | | | | | | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana 8622, Norway
| | | | | | - Obid Abdurakhmanovich Khamidov
- Rehabilitology and Sports Medicine Research Institute of the Samarkand State Medical University, Samarkand 140100, Uzbekistan
| | | |
Collapse
|
5
|
Yin Y, Liu Y, Du L, Wu S. Compromised B-cell homeostasis: Unraveling the link between major depression, infection and autoimmune disorders. J Affect Disord 2025; 374:565-578. [PMID: 39842671 DOI: 10.1016/j.jad.2025.01.095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 10/22/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Major depression can increase susceptibility to viral infections and autoimmune diseases. B cell responses are crucial for immune defense against infections but can trigger autoimmunity when deregulated. However, it remains unclear whether compromised B-cell homeostasis in major depression contributes to an increased risk of infection and autoimmunity. METHODS Chronic unpredictable mild stress (CUMS) procedure was applied to adult C57BL/6 J mice to generate a reliable depression model. Mice were immunized with (4-hydroxy-3-nitrophenyl) acetyl (NP) keyhole limpet hemocyanin (NP-KLH) to elicit B-cell-mediated humoral immune responses. CUMS mice were subjected to a collagen-induced arthritis model or a Bm12-induced systemic lupus erythematosus model to assess the contribution of major depression to autoimmunity. RNA sequencing was performed to understand the effects of CUMS on B-cell homeostasis at the transcriptomic level. RESULTS CUMS mice exhibited an impaired humoral immune response, as evidenced by reduced germinal centers (GCs), plasma cells, and antigen-specific antibodies. Unimmunized CUMS mice displayed aberrant spontaneous expansion of GC B cells, plasma cells, age-associated B cells and autoantibody production. CUMS mice also demonstrated a greater exacerbation of autoimmune manifestations. RNA sequencing revealed that genes involved in B-cell-mediated immune response were downregulated in B cells from CUMS mice, while the pathways related to autoimmunity seem to be upregulated. LIMITATIONS Further research is needed to understand the specific targets, mechanisms, and role of B cell dysfunction in major depression. CONCLUSIONS Our results provide novel insights into B-cell-dependent mechanisms that involve the association of increased susceptibility to infections and autoimmunity in major depression.
Collapse
Affiliation(s)
- Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuan Liu
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
6
|
Izuka S, Komai T, Itamiya T, Ota M, Nagafuchi Y, Shoda H, Matsuki K, Yamamoto K, Okamura T, Fujio K. Machine learning-driven immunophenotypic stratification of mixed connective tissue disease, corroborating the clinical heterogeneity. Rheumatology (Oxford) 2025; 64:1409-1416. [PMID: 38479808 PMCID: PMC11879315 DOI: 10.1093/rheumatology/keae158] [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: 01/04/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2025] Open
Abstract
OBJECTIVE The objective of this study was to stratify patients with MCTD, based on their immunophenotype. METHODS We analysed the immunophenotype and transcriptome of 24 immune cell subsets [from patients with MCTD, SLE, idiopathic inflammatory myopathy (IIM) and SSc] from our functional genome database, ImmuNexUT (https://www.immunexut.org/). MCTD patients were stratified by employing machine-learning models, including Random Forest, trained by immunophenotyping data from SLE, IIM and SSc patients. The transcriptomes were analysed with gene set variation analysis (GSVA), and the clinical features of the MCTD subgroups were compared. RESULTS This study included 215 patients, including 22 patients with MCTD. Machine-learning models, constructed to classify SLE, IIM and SSc patients, based on immunophenotyping, were applied to MCTD patients, resulting in 16 patients being classified as having an SLEimmunophenotype and 6 as having a non-SLE immunophenotype. Among the MCTD patients, patients with the SLE immunophenotype had higher proportions of Th1 cells {2.85% [interquartile range (IQR) 1.54-3.91] vs 1.33% (IQR 0.99-1.74) P = 0.027} and plasmablasts [6.35% (IQR 4.17-17.49) vs 2.00% (IQR 1.20-2.80) P = 0.010]. Notably, the number of SLE-related symptoms was higher in patients with the SLE immunophenotype [2.0 (IQR 1.0-2.0) vs 1.0 (IQR 1.0-1.0) P = 0.038]. Moreover, the GSVA scores of interferon-α and -γ responses were significantly higher in patients with the SLE immunophenotype in central memory CD8+ T cells, while hedgehog signalling was higher in patients with the non-SLE immunophenotype, in five-cell subsets. CONCLUSION This study describes the stratification of MCTD patients, based on immunophenotyping, suggesting the presence of distinct immunological processes behind the clinical subtypes of MCTD.
Collapse
Affiliation(s)
- Shinji Izuka
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshihiko Komai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahiro Itamiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kosuke Matsuki
- Research Division, Chugai Pharmaceutical Co., Ltd, Yokohama, Kanagawa, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Yu S, Zhou X, Liu R, Xu X, Ma D, Feng Y, Lin X. Immunomodulatory effects of Yu-Ping-Feng formula on primary Sjögren syndrome: interrogating the T-cell response. J Leukoc Biol 2025; 117:qiae155. [PMID: 38953166 DOI: 10.1093/jleuko/qiae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/23/2024] [Accepted: 07/01/2024] [Indexed: 07/03/2024] Open
Abstract
Ethnopharmacological treatments have shown beneficial effects in the clinical practice of autoimmune disorders. However, the underlying mechanism of immunomodulatory effects remains challenging, given the complicate composition of herbal medicines. Here, we developed an immunological approach to interrogate the T helper cell response. Through data mining, we hypothesized that Chinese medicine formula Yu-Ping-Feng might be a promising candidate for treating primary Sjögren syndrome, a common autoimmune disease manifested by exocrine gland dysfunction. We took advantage of a mouse model of experimental Sjögren syndrome that we previously established for Yu-Ping-Feng formula treatment. Yu-Ping-Feng therapy ameliorated the experimental Sjögren syndrome pathology in mice with active disease, showing improved salivary function and decreased serum levels of autoantibodies. Phenotypic analysis suggested that both effector T and B cells were significantly suppressed. Using coculture assay and adoptive transfer models, we demonstrated that Yu-Ping-Feng formula directly restrained effector/memory T-cell expansion and differentiation into Th17 and T follicular helper cells, the key subsets in experimental Sjögren syndrome pathogenesis. Importantly, we recruited 20 patients with primary Sjögren syndrome and conducted a pilot study of 8-wk therapy of Yu-Ping-Feng formula. Yu-Ping-Feng treatment effectively improved fatigue symptoms and exocrine gland functions, as well as reduced serum IgG/IgA levels, while effector T- and B-cell subsets were significantly decreased. There was a trend of reduction on disease activity, but not statistically significant. Together, our findings suggest a novel approach to assess the immunomodulatory effects of Yu-Ping-Feng formula, which may be favorable for patients with autoimmune disorders.
Collapse
Affiliation(s)
- Sulan Yu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hon Kong SAR 999077, China
| | - Xinyao Zhou
- Department of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijng 100053, China
| | - Ruihua Liu
- Department of Rheumatology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijng 100053, China
| | - Xiaoyu Xu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hon Kong SAR 999077, China
| | - Danbao Ma
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hon Kong SAR 999077, China
| | - Yun Feng
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China
| | - Xiang Lin
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hon Kong SAR 999077, China
- Department of Chinese Medicine, the University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen 518053, China
| |
Collapse
|
8
|
Fike AJ, Bricker KN, Gonzalez MV, Maharjan A, Bui T, Nuon K, Emrich SM, Weber JL, Luckenbill SA, Choi NM, Sauteraud R, Liu DJ, Olsen NJ, Caricchio R, Trebak M, Chodisetti SB, Rahman ZS. IRF7 controls spontaneous autoimmune germinal center and plasma cell checkpoints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.04.636277. [PMID: 39974943 PMCID: PMC11838595 DOI: 10.1101/2025.02.04.636277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
How IRF7 promotes autoimmune B cell responses and systemic autoimmunity is unclear. Analysis of spontaneous SLE-prone mice deficient in IRF7 uncovered the IRF7 role in regulating autoimmune germinal center (GC), plasma cell (PC) and autoantibody responses and disease. IRF7, however, was dispensable for foreign antigen driven GC, PC and antibody responses. Competitive bone marrow (BM) chimeras highlighted the importance of IRF7 in hematopoietic cells in spontaneous GC and PC differentiation. Single-cell-RNAseq of SLE-prone B cells indicated IRF7 mediated B cell differentiation through GC and PC fates. Mechanistic studies revealed that IRF7 promoted B cell differentiation through GC and PC fates by regulating the transcriptome, translation, and metabolism of SLE-prone B cells. Mixed BM chimeras demonstrated a requirement for B cell-intrinsic IRF7 in IgG autoantibody production but not sufficient for promoting spontaneous GC and PC responses. Altogether, we delineate previously unknown B cell-intrinsic and -extrinsic mechanisms of IRF7-promoted spontaneous GC and PC responses, loss of tolerance, autoantibody production and SLE development.
Collapse
Affiliation(s)
- Adam J. Fike
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Kristen N. Bricker
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Michael V. Gonzalez
- Center for Cytokine Storm Treatment and Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19464, USA
| | | | | | | | - Scott M. Emrich
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Julia L. Weber
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Sara A. Luckenbill
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Nicholas M. Choi
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Renan Sauteraud
- Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Dajiang J. Liu
- Public Health Science, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Nancy J. Olsen
- Rheumatology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | | | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Sathi Babu Chodisetti
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Ziaur S.M. Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| |
Collapse
|
9
|
Kono DH, Hahn BH. Animal models of systemic lupus erythematosus (SLE). DUBOIS' LUPUS ERYTHEMATOSUS AND RELATED SYNDROMES 2025:189-234. [DOI: 10.1016/b978-0-323-93232-5.00024-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Peroumal D, Jawale CV, Choi W, Rahimi H, Antos D, Li DD, Wang S, Manakkat Vijay GK, Mehta I, West R, Thangaraju M, Nolin TD, Das J, Alcorn JF, Biswas PS. The survival of B cells is compromised in kidney disease. Nat Commun 2024; 15:10842. [PMID: 39738044 PMCID: PMC11685736 DOI: 10.1038/s41467-024-55187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 12/04/2024] [Indexed: 01/01/2025] Open
Abstract
Antibody-mediated protection against pathogens is crucial to a healthy life. However, the recent SARS-CoV-2 pandemic has shown that pre-existing comorbid conditions including kidney disease account for compromised humoral immunity to infections. Individuals with kidney disease are not only susceptible to infections but also exhibit poor vaccine-induced antibody response. Using multiple mouse models of kidney disease, we demonstrate that renal dysfunction inhibits germinal center (GC) response against T-dependent antigens. GC B cells exhibit increased apoptosis in kidney disease. Uremic toxin hippuric acid drives loss of mitochondrial membrane potential, leading to increased apoptosis of GC B cells in a G-protein-coupled receptor 109A dependent manner. Finally, GC B cells and antibody titer are diminished in mice with kidney disease following influenza virus infection, a major cause of mortality in individuals with renal disorders. These results provide a mechanistic understanding of how renal dysfunction suppresses humoral immunity in patients with kidney disease.
Collapse
Affiliation(s)
- Doureradjou Peroumal
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chetan V Jawale
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wonseok Choi
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Hossein Rahimi
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Danielle Antos
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - De-Dong Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shuxia Wang
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Godhev K Manakkat Vijay
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, Department of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Isha Mehta
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, Department of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Raymond West
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Muthusamy Thangaraju
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Thomas D Nolin
- Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Systems Immunology, Department of Immunology and Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F Alcorn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Partha S Biswas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
11
|
Ding Q, Zhou Y, Feng Y, Sun L, Zhang T. Bruton's tyrosine kinase: A promising target for treating systemic lupus erythematosus. Int Immunopharmacol 2024; 142:113040. [PMID: 39216117 DOI: 10.1016/j.intimp.2024.113040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Systemic lupus erythematosus (SLE) is a chronic systemic autoimmune disorder involving multiple organs and systems. There is growing evidence that autoreactive B cells occupy a central role in the occurrence and progression of SLE due to their ability to generate pathogenic autoantibodies. Small molecule inhibitors targeting Bruton's tyrosine kinase (BTK), a crucial intracellular kinase regulating B cell development and function, emerge as a new strategy to treat SLE in recent years and are superior to biologic agents depleting B cells in many aspects. Supportive data obtained from lupus-prone mice preliminarily demonstrated the promising therapeutic potential of BTK inhibition. However, these BTK inhibitors, including elsubrutinib, evobrutinib, etc., mostly face with unsatisfactory efficacy and certain safety issues during clinical use, driving the quest for new-generation inhibitors with improved potency and higher selectivity. This paper elaborates the importance of BTK involvement in SLE pathogenesis, reviews the clinical research progress of BTK inhibitors for SLE and discusses limitations and challenges the drugs met in development, in order to contribute to a deeper understanding of disease mechanism and provide a reference for new-generation BTK inhibitor research.
Collapse
Affiliation(s)
- Qiaoyi Ding
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yifan Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lan Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
12
|
Li L, Huang H, Wang H, Pan Y, Tao H, Zhang S, Karmaus PWF, Fessler MB, Sleasman JW, Zhong XP. DGKα and ζ Deficiency Causes Regulatory T-Cell Dysregulation, Destabilization, and Conversion to Pathogenic T-Follicular Helper Cells to Trigger IgG1-Predominant Autoimmunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625360. [PMID: 39651265 PMCID: PMC11623591 DOI: 10.1101/2024.11.26.625360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Regulatory T cells (Tregs) actively engage in immune suppression to prevent autoimmune diseases but also inhibit anti-tumor immunity. Although Tregs express a TCR repertoire with relatively high affinities to self, they are normally quite stable and their inflammatory programs are intrinsically suppressed. We report here that diacylglycerol (DAG) kinases (DGK) ( and ( are crucial for homeostasis, suppression of proinflammatory programs, and stability of Tregs and for enforcing their dependence on CD28 costimulatory signal. Treg-specific deficiency of both DGK( and ( derails signaling, metabolic, and transcriptional programs in Tregs to cause dysregulated phenotypic and functional properties and to unleash conversion to pathogenic exTregs, especially exTreg-T follicular helper (Tfh) 2 cells, leading to uncontrolled effector T cell differentiation, deregulated germinal center (GC) B-cell responses and IgG1/IgE predominant antibodies/autoantibodies, and multiorgan autoimmune diseases. Our data not only illustrate the crucial roles of DGKs in Tregs to maintain self-tolerance but also unveil a Treg-to-self-reactive-pathogenic-exTreg-Tfh-cell program that is suppressed by DGKs and that could exert broad pathogenic roles in autoimmune diseases if unchecked.
Collapse
|
13
|
Jang E, Kim C, Noh J, Yi H, Jo S, Park JS, Hwang W, Cha JY, Cho ML, Kim TH, Youn J. Bach2 repression of CD36 regulates lipid-metabolism-linked effector functions in follicular B cells. Cell Rep 2024; 43:114878. [PMID: 39412989 DOI: 10.1016/j.celrep.2024.114878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/24/2024] [Accepted: 09/30/2024] [Indexed: 10/18/2024] Open
Abstract
The transcription repressor Bach2 plays a crucial role in shaping humoral immunity, but its cell-autonomous function remains elusive. Here, we reveal the mechanism by which Bach2 regulates effector cell maturation in peripheral B cells. In response to Toll-like receptor (TLR) agonists, Bach2 deficiency promotes the differentiation of follicular, but not marginal zone, B cells into effector cells, producing interleukin (IL)-6 and antibodies. This phenomenon is associated with changes in lipid metabolism, such as increases in CD36 expression, lipid influx, and fatty acid oxidation. Consistent with this, Bach2-deficient B cells exhibit elevated levels of mitochondrial oxidative stress, lipid peroxidation, and p38 activation. Mechanistically, Bach2 acts as a repressor of Cd36, and inhibition of CD36 or fatty acid oxidation reduces the differentiation of naive B cells into IL-6- and antibody-secreting cells. These results indicate Bach2 as a key metabolic checkpoint regulator crucial for maintaining a functionally quiescent state of follicular B cells.
Collapse
Affiliation(s)
- Eunkyeong Jang
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul 04763, Korea.
| | - ChangYeon Kim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Jeonghyun Noh
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Hansol Yi
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul 04763, Korea
| | - Jin-Sil Park
- Rheumatism Research Center, Catholic Institutes of Medical Science, The Catholic University of Korea, Seoul 06591, Korea
| | - Woochang Hwang
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea; Department of Pre-Medicine, College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Ji-Young Cha
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21936, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Institutes of Medical Science, The Catholic University of Korea, Seoul 06591, Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Hanyang University, Seoul 04763, Korea
| | - Jeehee Youn
- Laboratory of Autoimmunology, Department of Anatomy and Cell Biology, College of Medicine, Hanyang University, Seoul 04763, Korea; Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea.
| |
Collapse
|
14
|
Johansen ER, Schmalzriedt DL, Avila D, Sylvester PA, Rahlf CR, Bobek JM, Sahoo D, Dittel BN, Tarakanova VL. Combination of proviral and antiviral roles of B cell-intrinsic STAT1 expression defines parameters of chronic gammaherpesvirus infection. mBio 2024; 15:e0159824. [PMID: 39440973 PMCID: PMC11559066 DOI: 10.1128/mbio.01598-24] [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: 05/24/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Gammaherpesviruses are species-specific, ubiquitous pathogens that establish lifelong infection in their hosts and are associated with cancers, including B cell lymphomas. Type I and II interferons (IFNs) are critical for the control of acute and chronic gammaherpesvirus infection. However, the cell type-specific role of IFN signaling during natural infection is poorly defined and is masked by the altered viral pathogenesis observed in hosts with global IFN deficiencies. STAT1 is a constitutively expressed transcription factor that is critical for the effector function of type I and II IFNs. In this study, we defined the impact of B cell-specific STAT1 expression on gammaherpesvirus infection using murine gammaherpesvirus 68 (MHV68). While the acute stage of MHV68 infection was not affected, we found opposite, anatomic site-dependent effects of B cell-intrinsic STAT1 expression during chronic infection. Consistent with the antiviral role of STAT1, B cell-specific STAT1 expression attenuated the latent viral reservoir in peritoneal B cells of chronically infected mice. In contrast, STAT1 expression in splenic B cells supported the establishment of the latent MHV68 reservoir in germinal center B cells, revealing an unexpected proviral role of B cell-intrinsic STAT1 expression during chronic gammaherpesvirus infection. These STAT1-dependent MHV68 chronic infection phenotypes were fully recapitulated in the peritoneal cavity but not the spleen of mice with B cell-specific deficiency of type I IFN receptor. In summary, our study uncovers the intriguing combination of proviral and antiviral roles of B cell-intrinsic STAT1 expression during chronic gammaherpesvirus infection.IMPORTANCEInterferons (IFNs) execute broadly antiviral roles during acute and chronic viral infections. The constitutively expressed transcription factor STAT1 is a critical downstream effector of IFN signaling. Our studies demonstrate that B cell-intrinsic STAT1 expression has opposing and anatomic site-dependent roles during chronic gammaherpesvirus infection. Specifically, B cell-intrinsic STAT1 expression restricted gammaherpesvirus latent reservoir in the peritoneal cavity, consistent with the classical antiviral role of STAT1. In contrast, decreased STAT1 expression in splenic B cells led to the attenuated establishment of gammaherpesvirus latency and decreased latent infection of germinal center B cells, highlighting a novel proviral role of B cell-intrinsic STAT1 expression during chronic infection with a B cell-tropic gammaherpesvirus. Interestingly, B cell-specific type I IFN receptor deficiency primarily recapitulated the antiviral role of B cell-intrinsic STAT1 expression, suggesting the compensatory function of B cell-intrinsic type II IFN signaling or an IFN-independent proviral role of B cell-intrinsic STAT1 expression during chronic gammaherpesvirus infection.
Collapse
Affiliation(s)
- Erika R. Johansen
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Damon L. Schmalzriedt
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Danilela Avila
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Paul A. Sylvester
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Cade R. Rahlf
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jordan M. Bobek
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Daisy Sahoo
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Vera L. Tarakanova
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Cancer Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
15
|
Jinshan Z, Yong Q, Fangqi C, Juanmei C, Min L, Changzheng H. The role of TNF-α as a potential marker for acute cutaneous lupus erythematosus in patients with systemic lupus erythematosus. J Dermatol 2024; 51:1481-1491. [PMID: 38963308 DOI: 10.1111/1346-8138.17355] [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: 01/28/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
Acute cutaneous lupus erythematosus (ACLE) is closely associated with systemic symptoms in systemic lupus erythematosus (SLE). This study aimed to identify potential biomarkers for ACLE and explore their association with SLE to enable early prediction of ACLE and identify potential treatment targets for the future. In total, 185 SLE-diagnosed patients were enrolled and categorized into two groups: those with ACLE and those without cutaneous involvement. After conducting logistic regression analysis of the differentiating factors, we concluded that tumor necrosis factor-alpha (TNF-α) is an independent risk factor for ACLE. Analysis of the receiver operating characteristic revealed an area under the curve of 0.716 for TNF-α. Additionally, both TNF-α and ACLE are positively correlated with disease activity. TNF-α shows promise as a biomarker for ACLE, and in SLE patients, ACLE may serve as a clear indicator of moderate-to-severe disease activity.
Collapse
Affiliation(s)
- Zhan Jinshan
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qu Yong
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chen Fangqi
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Cao Juanmei
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
- Department of Dermatology, The First Affiliated Hospital of Shihezi University, Shihezi University, Shihezi, Xinjiang, China
| | - Li Min
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Huang Changzheng
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
16
|
Boudigou M, Frutoso M, Hémon P, Le Dantec C, Chatzis L, Devauchelle V, Jamin C, Cornec D, Pers JO, Le Pottier L, Hillion S. Phenotypic, transcriptomic, and spatial characterization of CD45RB + naïve mature B cells: Implications in Sjögren's disease. Clin Immunol 2024; 268:110378. [PMID: 39393568 DOI: 10.1016/j.clim.2024.110378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 10/13/2024]
Abstract
The conventional classification of mature B cells overlooks the diversity within IgD+ CD27- naïve B cells. Here, to identify distinct mature naïve B cells, we categorized CD45RBMEM55- B cells (NA RB-) and CD45RBMEM55+ B cells (NA RB+) and explore their function and localization in circulation and tissues under physiological and pathological conditions. NA RB+ B cells, found in secondary lymphoid organs, differentiate into plasmablasts and secrete IgM. In Sjögren's disease, their numbers decrease, and they show over-activation and abnormal migration, suggesting an adaptive disease response. NA RB+ B cells also appear in inflamed salivary glands, indicating involvement in local immune responses. These findings highlight the distinct roles of NA RB+ B cells in health and Sjögren's disease.
Collapse
Affiliation(s)
| | | | | | | | - Loukas Chatzis
- UMR1227, LBAI, Univ Brest, Inserm, Brest, France; Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Divi Cornec
- UMR1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France
| | | | | | - Sophie Hillion
- UMR1227, LBAI, Univ Brest, Inserm, and CHU Brest, Brest, France.
| |
Collapse
|
17
|
Moysidou E, Christodoulou M, Lioulios G, Stai S, Karamitsos T, Dimitroulas T, Fylaktou A, Stangou M. Lymphocytes Change Their Phenotype and Function in Systemic Lupus Erythematosus and Lupus Nephritis. Int J Mol Sci 2024; 25:10905. [PMID: 39456692 PMCID: PMC11508046 DOI: 10.3390/ijms252010905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease, characterized by considerable changes in peripheral lymphocyte structure and function, that plays a critical role in commencing and reviving the inflammatory and immune signaling pathways. In healthy individuals, B lymphocytes have a major role in guiding and directing defense mechanisms against pathogens. Certain changes in B lymphocyte phenotype, including alterations in surface and endosomal receptors, occur in the presence of SLE and lead to dysregulation of peripheral B lymphocyte subpopulations. Functional changes are characterized by loss of self-tolerance, intra- and extrafollicular activation, and increased cytokine and autoantibody production. T lymphocytes seem to have a supporting, rather than a leading, role in the disease pathogenesis. Substantial aberrations in peripheral T lymphocyte subsets are evident, and include a reduction of cytotoxic, regulatory, and advanced differentiated subtypes, together with an increase of activated and autoreactive forms and abnormalities in follicular T cells. Up-regulated subpopulations, such as central and effector memory T cells, produce pre-inflammatory cytokines, activate B lymphocytes, and stimulate cell signaling pathways. This review explores the pivotal roles of B and T lymphocytes in the pathogenesis of SLE and Lupus Nephritis, emphasizing the multifaceted mechanisms and interactions and their phenotypic and functional dysregulations.
Collapse
Affiliation(s)
- Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Theodoros Karamitsos
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Cardiology, AHEPA University Hospital, 54636 Thessaloniki, Greece
| | - Theodoros Dimitroulas
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 4th Department of Medicine, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece;
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.M.); (M.C.); (G.L.); (S.S.); (T.K.); (T.D.)
- 1st Department of Nephrology, Hippokration General Hospital, 54642 Thessaloniki, Greece
| |
Collapse
|
18
|
Dey D, Biswas S, Pal S, Nandi S, Khatun N, Jha R, Chakraborty BC, Baidya A, Ghosh R, Banerjee S, Ahammed SKM, Chowdhury A, Datta S. Monocyte-derived Galectin-9 and PD-L1 differentially impair adaptive and innate immune response in chronic HBV infection and their expression remain unaltered after antiviral therapy. Front Immunol 2024; 15:1474853. [PMID: 39445017 PMCID: PMC11496065 DOI: 10.3389/fimmu.2024.1474853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Patients with chronic HBV infection (CHI) exhibit defective anti-viral immune-response whose underlying causes still remain unclear. Monocytes act as immune sentinels for pathogens and can regulate immunity via interaction with other immune-cells, apart from differentiating into macrophages. Immune-checkpoint molecules (ICMs) expressed by immune-cells, including monocytes are known to negatively regulate immune-responses. Here, we evaluated the expression of ICMs, namely, Gal-9, PD-L1, and CTLA-4 on monocytes in different phases of CHI, identified the viral and the host factors causing their aberrant expression and investigated their impact during interaction of monocytes with T-cells, B-cells and NK-cells and also on monocyte to macrophage differentiation. Influence of Tenofovir therapy on the expression of monocytic ICMs was also studied. Methods Collection of blood and liver-tissue samples from HBV infected patients and controls, flow-cytometry, cell sorting, cell culture and immune-fluorescence were performed for this study. Results Gal-9+ and PD-L1+-monocytes were significantly increased in HBeAg-positive as well as HBeAg-negative chronic hepatitis B (CHB) patients than healthy controls (HC). In immune-tolerant (IT) subjects, only Gal-9+-monocytes and in inactive carriers (IC), PD-L1+-monocytes were higher than HC while CTLA-4+-monocytes remained comparable among groups. High serum Hepatitis B surface antigen (HBsAg) concentration in CHB as well as IT and TNF-α in CHB triggered monocytic Gal-9-expression whereas, PD-L1 was induced by elevated TNF-α and IL-4 in CHB and IL-1β in CHB and IC. Purified monocytes from CHB and IT having high Gal-9 expression led to expansion of CD4+CD25+FOXP3+-Tregs, CD19+IL-10+-Bregs and CD19+CD27-CD21-atypical memory B-cells and these monocytes also preferentially differentiated into M2-macrophages. These phenomena were reversed by anti-Gal-9-antibody. Parallelly, PD-L1+-monocytes in CHB and IC reduced IL-2/IFN-γ and IL-6 production by HBV-specific T- and B-cells respectively, which were restored by anti-PD-L1-antibody. Both Gal-9+- and PD-L1+-monocytes caused decline in IFN-γ+-NK-cells but enhanced IL-10-expressing HBV-specific-T-cells and NK-cells. Increased intrahepatic CD14+Gal-9+ and CD14+PD-L1+-monocytes were noted in CHB patients than HC. One-year tenofovir therapy failed to reduce monocytic Gal-9 and PD-L1 along with the levels of HBsAg, TNF-α, IL-1β and IL-4. Conclusions Monocytic Gal-9 and PD-L1, expressed heterogeneously in different phases of CHI, exert diverse inhibitory effects on immune-responses and their therapeutic targeting could boost anti-HBV immunity.
Collapse
Affiliation(s)
- Debangana Dey
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Satabdi Biswas
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Sourina Pal
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Sarthak Nandi
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Najma Khatun
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Rambha Jha
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Bidhan Chandra Chakraborty
- Multidisciplinary Research Unit, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Ayana Baidya
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Ranajoy Ghosh
- Division of Pathology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Soma Banerjee
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - SK Mahiuddin Ahammed
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Abhijit Chowdhury
- Department of Hepatology, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| | - Simanti Datta
- Centre for Liver Research, School of Digestive and Liver Diseases, Institute of Post Graduate Medical Education and Research, Kolkata, India
| |
Collapse
|
19
|
Ortiz G, Blanco T, Singh RB, Kahale F, Wang S, Chen Y, Dana R. IL-6 induces Treg dysfunction in desiccating stress-induced dry eye disease. Exp Eye Res 2024; 246:110006. [PMID: 39009059 PMCID: PMC11332651 DOI: 10.1016/j.exer.2024.110006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/03/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Regulatory T cells (Tregs) play a critical role in maintaining immune homeostasis, and their dysfunction is implicated in the pathogenesis of various autoimmune disorders, including dry eye disease (DED). Treg dysfunction in DED allows T-helper cell 17 (Th17) mediated chronic inflammation at the ocular surface. In this study, the factors causing Treg dysfunction in DED were investigated. We observed reduced expression of Treg functional markers - FoxP3, CD25, and CTLA-4 in the cells isolated from DED mice (DED Tregs). Additionally, DED Tregs showed increased expression levels of receptors for pro-inflammatory cytokine receptors, namely IL-6R, IL-17RA, and IL-23R. An increased expression level of pro-inflammatory cytokine receptors was observed on exposing Tregs isolated from naïve mice (NTregs) to IL-6 or IL-17, but not IL-23, with a concomitant downregulation of FoxP3, CD25, and CTLA-4 in these cells. Furthermore, among these cytokines, IL-6 induced the most pronounced loss of Treg mediated suppression of Th17 proliferation and IL-10 secretion. In vitro and in vivo blockade of IL-6 effectively restored function in DED Tregs, leading to enhanced suppressive function against proliferating Th17 cells and ameliorating disease severity. In conclusion, this study provides insights into mechanisms of Treg dysregulation in DED, specifically delineating the effect of Th17-associated cytokines, with IL-6 emerging as the critical factor inducing Treg dysfunctionality. These findings highlight the potential for developing novel therapeutic interventions for DED through restoration of immunosuppressive function of Tregs.
Collapse
Affiliation(s)
- Gustavo Ortiz
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tomas Blanco
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Rohan Bir Singh
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Francesca Kahale
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Shudan Wang
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Yihe Chen
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Niu L, Chen T, Yang A, Yan X, Jin F, Zheng A, Song X. Macrophages and tertiary lymphoid structures as indicators of prognosis and therapeutic response in cancer patients. Biochim Biophys Acta Rev Cancer 2024; 1879:189125. [PMID: 38851437 DOI: 10.1016/j.bbcan.2024.189125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Tertiary lymphoid structures (TLS) can reflect cancer prognosis and clinical outcomes in various tumour tissues. Tumour-associated macrophages (TAMs) are indispensable components of the tumour microenvironment and play crucial roles in tumour development and immunotherapy. TAMs are associated with TLS induction via the modulation of the T cell response, which is a major component of the TLS. Despite their important roles in cancer immunology, the subtypes of TAMs that influence TLS and their correlation with prognosis are not completely understood. Here, we provide novel insights into the role of TAMs in regulating TLS formation. Furthermore, we discuss the prognostic value of these TAM subtypes and TLS, as well as the current antitumour therapies for inducing TLS. This study highlights an entirely new field of TLS regulation that may lead to the development of an innovative perspective on immunotherapy for cancer treatment.
Collapse
Affiliation(s)
- Li Niu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Aodan Yang
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Xiwen Yan
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China
| | - Ang Zheng
- Department of Breast Surgery, the First Hospital of China Medical University, Shenyang, China.
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Wu Y, Zhou Y, Zhu Q, Liu Y, Deng D, Zhang J. B cell function in patients with systemic lupus erythematosus is regulated by the upregulation of JunD. Heliyon 2024; 10:e35949. [PMID: 39220919 PMCID: PMC11365431 DOI: 10.1016/j.heliyon.2024.e35949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose Systemic lupus erythematosus (SLE) is largely caused by B cell dysfunction. JunD is an activator protein 1 family protein that has been linked to the regulation of apoptotic and proliferative activities. However, the precise mechanism(s) by which JunD functions remains to be fully elucidated. Accordingly, this study aimed to clarify the functional importance of JUND gene expression in SLE, with further analyses of the functional role that JunD plays as a regulator of B cell proliferation and immune function. Methods Reverse transcriptase quantitative polymerase chain reaction techniques were used to analyze JunD expression in B cells of patients with SLE and healthy subjects. Cell Counting Kit-8 (CCK-8) assays and flow cytometry methods were used to characterise proliferative activity, cell cycle progression, and apoptosis of B cells in which JunD was either knocked down or overexpressed. The immune status and autophagic activity of these cells were assessed using Western immunoblotting and enzyme-linked immunosorbent assay (ELISA). Additionally, a JunD knockdown mouse model was established, and the functional role of B cell JunD expression in the pathogenesis of SLE was assessed using Western immunoblotting, ELISA, and haematoxylin and eosin staining. Results B cells from patients with SLE exhibited upregulation of JunD, with overexpression facilitating in vitro cellular proliferation and modulation of the immune and autophagic status of these B cells. JunD knockdown was also sufficient to modulate in vivo immune function and the autophagic status of B cells. Conclusion JunD was upregulated in the B cells of patients with SLE, where it regulates proliferation, autophagy, and immunity.
Collapse
Affiliation(s)
- Yongzhuo Wu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Yali Zhou
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Qinghuan Zhu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Yingying Liu
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Danqi Deng
- Department of Dermatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, PR China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing, 100044, PR China
| |
Collapse
|
22
|
Tsai PJ, Chen MY, Hsu WC, Lin SF, Chan PC, Chen HH, Kao CY, Lin WJ, Chuang TH, Yu GY, Su YW. PTEN acts as a crucial inflammatory checkpoint controlling TLR9/IL-6 axis in B cells. iScience 2024; 27:110388. [PMID: 39092178 PMCID: PMC11292540 DOI: 10.1016/j.isci.2024.110388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
Phosphatase and tensin homolog (PTEN) is vital for B cell development, acting as a key negative regulator in the PI3K signaling pathway. We used CD23-cre to generate PTEN-conditional knockout mice (CD23-cKO) to examine the impact of PTEN mutation on peripheral B cells. Unlike mb1-cre-mediated PTEN deletion in early B cells, CD23-cKO mutants exhibited systemic inflammation with increased IL-6 production in mature B cells upon CpG stimulation. Inflammatory B cells in CD23-cKO mice showed elevated phosphatidylinositol 3-phosphate [PI(3)P] levels and increased TLR9 endosomal localization. Pharmacological inhibition of PI(3)P synthesis markedly reduced TLR9-mediated IL-6. Single-cell RNA-sequencing (RNA-seq) revealed altered endocytosis, BANK1, and NF-κB1 expression in PTEN-deficient B cells. Ectopic B cell receptor (BCR) expression on non-inflammatory mb1-cKO B cells restored BANK1 and NF-κB1 expression, enhancing TLR9-mediated IL-6 production. Our study highlights PTEN as a crucial inflammatory checkpoint, regulating TLR9/IL-6 axis by fine-tuning PI(3)P homeostasis. Additionally, BCR downregulation prevents the differentiation of inflammatory B cells in PTEN deficiency.
Collapse
Affiliation(s)
- Pei-Ju Tsai
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Ming-Yu Chen
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Wei-Chan Hsu
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Su-Fang Lin
- National Institute of Cancer Research, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Po-Chiang Chan
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Hsin-Hsin Chen
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Cheng-Yuan Kao
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Wen-Jye Lin
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Guann-Yi Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Zhunan Town, Miaoli County 350401, Taiwan
| |
Collapse
|
23
|
Netti GS, Soccio P, Catalano V, De Luca F, Khalid J, Camporeale V, Moriondo G, Papale M, Scioscia G, Corso G, Foschino MP, Lo Caputo S, Lacedonia D, Ranieri E. The Onset of Antinuclear Antibodies (ANAs) as a Potential Risk Factor for Mortality and Morbidity in COVID-19 Patients: A Single-Center Retrospective Study. Biomedicines 2024; 12:1306. [PMID: 38927513 PMCID: PMC11201662 DOI: 10.3390/biomedicines12061306] [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: 05/13/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The immune system's amplified response to SARS-CoV-2 may lead to the production of autoantibodies, but their specific impact on disease severity and outcome remains unclear. This study aims to assess if hospitalized COVID-19 patients face a worse prognosis based on ANA presence, even without autoimmune diseases. We performed a retrospective, single-center, observational cohort study, enrolling 638 COVID-19 patients hospitalized from April 2020 to March 2021 at Hospital "Policlinico Riuniti" of Foggia (Italy). COVID-19 patients with a positive ANA test exhibited a significantly lower 30-day survival rate (64.4% vs. 83.0%) and a higher likelihood of severe respiratory complications during hospitalization than those with negative ANA screening (35.4% vs. 17.0%) (p < 0.001). The association between poor prognosis and ANA status was identified by calculating the HALP score (Hemoglobin-Albumin-Lymphocyte-Platelet), which was lower in COVID-19 patients with a positive ANA test compared to ANA-negative patients (108.1 ± 7.4 vs. 218.6 ± 11.2 AU; p < 0.011). In detail, COVID-19 patients with a low HALP showed a lower 30-day survival rate (99.1% vs. 83.6% vs. 55.2% for high, medium, and low HALP, respectively; p < 0.001) and a higher incidence of adverse respiratory events compared to those with high and medium HALP (13.1% vs. 35.2% vs. 64.6% for high, medium, and low HALP, respectively; p < 0.001). In summary, ANA positivity in COVID-19 patients appears to be linked to a more aggressive disease phenotype with a reduced survival rate. Furthermore, we propose that the HALP score could serve as a valuable parameter to assess prognosis for COVID-19 patients.
Collapse
Affiliation(s)
- Giuseppe Stefano Netti
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| | - Piera Soccio
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Valeria Catalano
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Federica De Luca
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Javeria Khalid
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Valentina Camporeale
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Giorgia Moriondo
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Massimo Papale
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| | - Giulia Scioscia
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Gaetano Corso
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
- Clinical Biochemistry, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Maria Pia Foschino
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Sergio Lo Caputo
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Donato Lacedonia
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Elena Ranieri
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| |
Collapse
|
24
|
Guo S, Tian Y, Li J, Zeng X. A Glimpse into Humoral Response and Related Therapeutic Approaches of Takayasu's Arteritis. Int J Mol Sci 2024; 25:6528. [PMID: 38928233 PMCID: PMC11203527 DOI: 10.3390/ijms25126528] [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: 05/04/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Takayasu's arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing body of evidence bears testimony to the widespread effects of B cells on disease pathogenesis and progression. Distinct alterations in peripheral B cell subsets have been described in individuals with TAK. Advancements in technology have facilitated the identification of novel autoantibodies in TAK. Moreover, emerging data suggest that dysregulated signaling cascades downstream of B cell receptor families, including interactions with innate pattern recognition receptors such as toll-like receptors, as well as co-stimulatory molecules like CD40, CD80 and CD86, may result in the selection and proliferation of autoreactive B cell clones in TAK. Additionally, ectopic lymphoid neogenesis within the aortic wall of TAK patients exhibits functional characteristics. In recent decades, therapeutic interventions targeting B cells, notably utilizing the anti-CD20 monoclonal antibody rituximab, have demonstrated efficacy in TAK. Despite the importance of the humoral immune response, a systematic understanding of how autoreactive B cells contribute to the pathogenic process is still lacking. This review provides a comprehensive overview of the biological significance of B cell-mediated autoimmunity in TAK pathogenesis, as well as insights into therapeutic strategies targeting the humoral response. Furthermore, it examines the roles of T-helper and T follicular helper cells in humoral immunity and their potential contributions to disease mechanisms. We believe that further identification of the pathogenic role of autoimmune B cells and the underlying regulation system will lead to deeper personalized management of TAK patients. We believe that further elucidation of the pathogenic role of autoimmune B cells and the underlying regulatory mechanisms holds promise for the development of personalized approaches to managing TAK patients.
Collapse
Affiliation(s)
- Shuning Guo
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Yixiao Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Jing Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100006, China; (S.G.); (Y.T.)
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Beijing 100006, China
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Beijing 100006, China
- Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100006, China
| |
Collapse
|
25
|
Doctor GT, Dudreuilh C, Perera R, Dorling A. Granulomatous Tubulointerstitial Nephritis in a Kidney Allograft: Treatment with Interleukin-6 Receptor Antagonist Stabilises Kidney Function. J Clin Med 2024; 13:3427. [PMID: 38929956 PMCID: PMC11205090 DOI: 10.3390/jcm13123427] [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: 04/08/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Granulomatous tubulointerstitial nephritis (GTIN) attributed to early onset sarcoidosis is an ultrarare finding in an allograft kidney biopsy. We present the case of a young man with allograft dysfunction who had GTIN upon biopsy. We performed a thorough case review based on recovered records from early childhood and reassessed genetic testing results. We revised his underlying diagnosis from cryopyrin-associated periodic syndrome to early-onset sarcoidosis with wild-type NOD2 and established a rationale to use the interleukin-6 (IL-6) receptor blocker tocilizumab (TCZ). This suppressed his inflammatory disease and stabilised kidney function. We performed a literature review related to the emerging role of IL-6 pathway blockade in kidney transplantation. We identified 18 reports with 417 unique patients treated with TCZ for indications including HLA-desensitisation, transplant immunosuppression induction, treatment of chronic antibody-mediated rejection, and treatment of subclinical rejection. Both TCZ and the direct IL-6 inhibitor clazakizumab are being studied in ongoing randomised control trials.
Collapse
Affiliation(s)
- Gabriel T. Doctor
- Department of Transplantation, Renal and Urology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (C.D.); (R.P.); (A.D.)
| | | | | | | |
Collapse
|
26
|
Nian Z, Mao Y, Xu Z, Deng M, Xu Y, Xu H, Chen R, Xu Y, Huang N, Mao F, Xu C, Wang Y, Niu M, Chen A, Xue X, Zhang H, Guo G. Multi-omics analysis uncovered systemic lupus erythematosus and COVID-19 crosstalk. Mol Med 2024; 30:81. [PMID: 38862942 PMCID: PMC11167821 DOI: 10.1186/s10020-024-00851-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Studies have highlighted a possible crosstalk between the pathogeneses of COVID-19 and systemic lupus erythematosus (SLE); however, the interactive mechanisms remain unclear. We aimed to elucidate the impact of COVID-19 on SLE using clinical information and the underlying mechanisms of both diseases. METHODS RNA-seq datasets were used to identify shared hub gene signatures between COVID-19 and SLE, while genome-wide association study datasets were used to delineate the interaction mechanisms of the key signaling pathways. Finally, single-cell RNA-seq datasets were used to determine the primary target cells expressing the shared hub genes and key signaling pathways. RESULTS COVID-19 may affect patients with SLE through hematologic involvement and exacerbated inflammatory responses. We identified 14 shared hub genes between COVID-19 and SLE that were significantly associated with interferon (IFN)-I/II. We also screened and obtained four core transcription factors related to these hub genes, confirming the regulatory role of the IFN-I/II-mediated Janus kinase/signal transducers and activators of transcription (JAK-STAT) signaling pathway on these hub genes. Further, SLE and COVID-19 can interact via IFN-I/II and IFN-I/II receptors, promoting the levels of monokines, including interleukin (IL)-6/10, tumor necrosis factor-α, and IFN-γ, and elevating the incidence rate and risk of cytokine release syndrome. Therefore, in SLE and COVID-19, both hub genes and core TFs are enriched within monocytes/macrophages. CONCLUSIONS The interaction between SLE and COVID-19 promotes the activation of the IFN-I/II-triggered JAK-STAT signaling pathway in monocytes/macrophages. These findings provide a new direction and rationale for diagnosing and treating patients with SLE-COVID-19 comorbidity.
Collapse
Affiliation(s)
- Zekai Nian
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yicheng Mao
- Ophthalmology College, Wenzhou Medical University, Wenzhou, China
| | - Zexia Xu
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ming Deng
- Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Yixi Xu
- School of Public Administration, Hangzhou Normal University, Hangzhou, China
| | - Hanlu Xu
- Ophthalmology College, Wenzhou Medical University, Wenzhou, China
| | - Ruoyao Chen
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Yiliu Xu
- Research Center of Fluid Machinery Engineering and Technology, Jiangsu University, Zhenjiang, China
| | - Nan Huang
- Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Feiyang Mao
- Second Clinical College, Wenzhou Medical University, Wenzhou, China
| | - Chenyu Xu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yulin Wang
- Public Health and Management College, Wenzhou Medical University, Wenzhou, China
| | - Mengyuan Niu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Aqiong Chen
- Department of Rheumatology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Huidi Zhang
- Department of Nephrology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Gangqiang Guo
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-Related Pathogens and Immunity, Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
27
|
Lovell CD, Jiwrajka N, Amerman HK, Cancro MP, Anguera MC. Xist Deletion in B Cells Results in Systemic Lupus Erythematosus Phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594175. [PMID: 38798403 PMCID: PMC11118349 DOI: 10.1101/2024.05.15.594175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease preferentially observed in females. X-linked gene expression in XX females is normalized to that of XY males by X-Chromosome Inactivation (XCI). However, B cells from female SLE patients and mouse models of SLE exhibit mislocalization of Xist RNA, a critical regulator of XCI, and aberrant expression of X-linked genes, suggesting that impairment of XCI may contribute to disease. Here, we find that a subset of female mice harboring a conditional deletion of Xis t in B cells ("Xist cKO") spontaneously develop SLE phenotypes, including expanded activated B cell subsets, disease-specific autoantibodies, and glomerulonephritis. Moreover, pristane-induced SLE-like disease is more severe in Xist cKO mice. Activated B cells from Xist cKO mice with SLE phenotypes have increased expression of proinflammatory X-linked genes implicated in SLE. Together, this work indicates that impaired XCI maintenance in B cells directly contributes to the female-bias of SLE.
Collapse
|
28
|
Dadgar N, Sherry C, Zimmerman J, Park H, Lewis C, Donnenberg A, Zaidi AH, Fan Y, Xiao K, Bartlett D, Donnenberg V, Wagner PL. Targeting interleukin-6 as a treatment approach for peritoneal carcinomatosis. J Transl Med 2024; 22:402. [PMID: 38689325 PMCID: PMC11061933 DOI: 10.1186/s12967-024-05205-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Peritoneal carcinomatosis (PC) is a complex manifestation of abdominal cancers, with a poor prognosis and limited treatment options. Recent work identifying high concentrations of the cytokine interleukin-6 (IL-6) and its soluble receptor (sIL-6-Rα) in the peritoneal cavity of patients with PC has highlighted this pathway as an emerging potential therapeutic target. This review article provides a comprehensive overview of the current understanding of the potential role of IL-6 in the development and progression of PC. We discuss mechansims by which the IL-6 pathway may contribute to peritoneal tumor dissemination, mesothelial adhesion and invasion, stromal invasion and proliferation, and immune response modulation. Finally, we review the prospects for targeting the IL-6 pathway in the treatment of PC, focusing on common sites of origin, including ovarian, gastric, pancreatic, colorectal and appendiceal cancer, and mesothelioma.
Collapse
Affiliation(s)
- Neda Dadgar
- Translational Hematology & Oncology Research, Enterprise Cancer Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Christopher Sherry
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Jenna Zimmerman
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Hyun Park
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Catherine Lewis
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Albert Donnenberg
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Ali H Zaidi
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Yong Fan
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Kunhong Xiao
- Center for Proteomics & Artificial Intelligence, Center for Clinical Mass Spectrometry, Allegheny Health Network Cancer Institute, Pittsburgh, PA, 15224, USA
| | - David Bartlett
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA
| | - Vera Donnenberg
- University of Pittsburgh School of MedicineDepartment of Cardiothoracic SurgeryUPMC Hillman Cancer Center Wagner, Patrick; Allegheny Health Network Cancer Institute, Pittsburgh, USA
| | - Patrick L Wagner
- Allegheny Health Network Cancer Institute, 314 E. North Ave, Pittsburgh, PA, 15212, USA.
| |
Collapse
|
29
|
Chen W, Toda E, Takeuchi K, Sawa Y, Wakamatsu K, Kuwahara N, Ishikawa A, Igarashi Y, Terasaki M, Kunugi S, Terasaki Y, Yamada K, Terashima Y, Shimizu A. Disulfiram treatment suppresses antibody-producing reactions by inhibiting macrophage activation and B cell pyrimidine metabolism. Commun Biol 2024; 7:488. [PMID: 38649462 PMCID: PMC11035657 DOI: 10.1038/s42003-024-06183-9] [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: 07/18/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Antibody responses, involving B cells, CD4 + T cells, and macrophages, are implicated in autoimmune diseases and organ transplant rejection. We have previously shown that inhibiting FROUNT with disulfiram (DSF) suppresses macrophage activation and migration, effectively treating inflammatory diseases. In this study, we investigated the effectiveness of DSF in antibody-producing reactions. Using a heart transplantation mouse model with antibody-mediated rejection, we administered anti-CD8 antibody to exclude cellular rejection. DSF directly inhibited B cell responses in vitro and significantly reduced plasma donor-specific antibodies and graft antibody deposition in vivo, resulting in prolonged survival of the heart graft. DSF also mediated various effects, including decreased macrophage infiltration and increased Foxp3+ regulatory T-cells in the grafts. Additionally, DSF inhibited pyrimidine metabolism-related gene expression induced by B-cell stimulation. These findings demonstrate that DSF modulates antibody production in the immune response complexity by regulating B-cell and macrophage responses.
Collapse
Affiliation(s)
- Weili Chen
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Etsuko Toda
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan.
- Laboratory for Morphological and Biomolecular Imaging, Nippon Medical School, Tokyo, Japan.
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan.
| | - Kazuhiro Takeuchi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Yurika Sawa
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Kyoko Wakamatsu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Naomi Kuwahara
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Arimi Ishikawa
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Yuri Igarashi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Shinobu Kunugi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | | | - Kazuhiko Yamada
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Yuya Terashima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan.
| |
Collapse
|
30
|
Flippot R, Teixeira M, Rey-Cardenas M, Carril-Ajuria L, Rainho L, Naoun N, Jouniaux JM, Boselli L, Naigeon M, Danlos FX, Escudier B, Scoazec JY, Cassard L, Albiges L, Chaput N. B cells and the coordination of immune checkpoint inhibitor response in patients with solid tumors. J Immunother Cancer 2024; 12:e008636. [PMID: 38631710 PMCID: PMC11029261 DOI: 10.1136/jitc-2023-008636] [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: 03/31/2024] [Indexed: 04/19/2024] Open
Abstract
Immunotherapy profoundly changed the landscape of cancer therapy by providing long-lasting responses in subsets of patients and is now the standard of care in several solid tumor types. However, immunotherapy activity beyond conventional immune checkpoint inhibition is plateauing, and biomarkers are overall lacking to guide treatment selection. Most studies have focused on T cell engagement and response, but there is a growing evidence that B cells may be key players in the establishment of an organized immune response, notably through tertiary lymphoid structures. Mechanisms of B cell response include antibody-dependent cellular cytotoxicity and phagocytosis, promotion of CD4+ and CD8+ T cell activation, maintenance of antitumor immune memory. In several solid tumor types, higher levels of B cells, specific B cell subpopulations, or the presence of tertiary lymphoid structures have been associated with improved outcomes on immune checkpoint inhibitors. The fate of B cell subpopulations may be widely influenced by the cytokine milieu, with versatile roles for B-specific cytokines B cell activating factor and B cell attracting chemokine-1/CXCL13, and a master regulatory role for IL-10. Roles of B cell-specific immune checkpoints such as TIM-1 are emerging and could represent potential therapeutic targets. Overall, the expanding field of B cells in solid tumors of holds promise for the improvement of current immunotherapy strategies and patient selection.
Collapse
Affiliation(s)
- Ronan Flippot
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Marcus Teixeira
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Macarena Rey-Cardenas
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Lucia Carril-Ajuria
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
- Medical Oncology, CHU Brugmann, Brussels, Belgium
| | - Larissa Rainho
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Natacha Naoun
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Jean-Mehdi Jouniaux
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Lisa Boselli
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Marie Naigeon
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Francois-Xavier Danlos
- LRTI, INSERM U1015, Gustave Roussy, Villejuif, France
- Drug Development Department, Gustave Roussy, Villejuif, France
| | - Bernard Escudier
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | | | - Lydie Cassard
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| | - Nathalie Chaput
- Immunomonitoring Laboratory, CNRS3655 & INSERM US23, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
31
|
Liu S, Lagos J, Shumlak NM, Largent AD, Lewis ST, Holder U, Du SW, Liu Y, Hou B, Acharya M, Jackson SW. NADPH oxidase exerts a B cell-intrinsic contribution to lupus risk by modulating endosomal TLR signals. J Exp Med 2024; 221:e20230774. [PMID: 38442270 PMCID: PMC10913815 DOI: 10.1084/jem.20230774] [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: 05/08/2023] [Revised: 11/11/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Abstract
Genome-wide association studies in systemic lupus erythematosus (SLE) have linked loss-of-function mutations in phagocytic NADPH oxidase complex (NOX2) genes, including NCF1 and NCF2, to disease pathogenesis. The prevailing model holds that reduced NOX2 activity promotes SLE via defective efferocytosis, the immunologically silent clearance of apoptotic cells. Here, we describe a parallel B cell-intrinsic mechanism contributing to breaks in tolerance. In keeping with an important role for B cell Toll-like receptor (TLR) pathways in lupus pathogenesis, NOX2-deficient B cells exhibit enhanced signaling downstream of endosomal TLRs, increased humoral responses to nucleic acid-containing antigens, and the propensity toward humoral autoimmunity. Mechanistically, TLR-dependent NOX2 activation promotes LC3-mediated maturation of TLR-containing endosomes, resulting in signal termination. CRISPR-mediated disruption of NCF1 confirmed a direct role for NOX2 in regulating endosomal TLR signaling in primary human B cells. Together, these data highlight a new B cell-specific mechanism contributing to autoimmune risk in NCF1 and NCF2 variant carriers.
Collapse
Affiliation(s)
- Shuozhi Liu
- Seattle Children’s Research Institute, Seattle, WA, USA
| | | | | | | | | | - Ursula Holder
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Samuel W. Du
- Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yifan Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Baidong Hou
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Mridu Acharya
- Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Shaun W. Jackson
- Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
32
|
Yang JH, Miner AE, Fair A, Kinkel R, Graves JS. Senescence marker p16INK4a expression in patients with multiple sclerosis. Mult Scler Relat Disord 2024; 84:105498. [PMID: 38359693 DOI: 10.1016/j.msard.2024.105498] [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: 10/23/2023] [Revised: 01/18/2024] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVES Telomere attrition is associated with disability accumulation and brain atrophy in multiple sclerosis (MS). Downstream of telomere attrition is cellular senescence. We sought to determine differences in the cellular senescence marker p16INK4a expression between MS and healthy control participants and the association of p16INK4a expression with MS disability and treatment exposure. METHODS Patients meeting diagnostic criteria for MS and healthy controls were recruited for a cross-sectional pilot study. RNA was extracted from peripheral blood mononuclear cells (PBMCs) and p16INK4a expression levels were measured using qRT PCR. Spearman correlation coefficients and regression models were applied to compare expression levels to chronological age, assess case control differences, and determine associations with clinical outcome measures. RESULTS Fifty-two participants with MS (67 % female, ages 25-70) and 38 healthy controls (66 % female, ages 23-65) were included. p16INK4a levels were not linearly correlated with chronological age in MS (rhos = -0.01, p = 0.94) or control participants (rhos = 0.02, p = 0.92). Higher median p16INK4a levels were observed in the >50-year age group for MS (0.25, IQR 0.14-0.35) vs. controls (0.12, IQR 0.05-0.15) and in this age group B cell depletion therapy was associated with lower expression levels. p16INK4a expression was not associated with any of the measured MS disability outcomes. DISCUSSION Caution is needed with using p16INK4a expression level from PBMCs as an aging biomarker in MS participants, given lack of correlation with chronological age or large associations with clinical outcomes.
Collapse
Affiliation(s)
- Jennifer H Yang
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA; Rady Children's Hospital San Diego, San Diego, CA, USA.
| | - Annalise E Miner
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA; Boston University, Boston, CA, USA
| | - Ashley Fair
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Revere Kinkel
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA; Rady Children's Hospital San Diego, San Diego, CA, USA
| |
Collapse
|
33
|
Zhu X, Hong S, Bu J, Liu Y, Liu C, Li R, Zhang T, Zhang Z, Li L, Zhou X, Hua Z, Zhu B, Hou B. Antiviral memory B cells exhibit enhanced innate immune response facilitated by epigenetic memory. SCIENCE ADVANCES 2024; 10:eadk0858. [PMID: 38552009 PMCID: PMC10980274 DOI: 10.1126/sciadv.adk0858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
The long-lasting humoral immunity induced by viral infections or vaccinations depends on memory B cells with greatly increased affinity to viral antigens, which are evolved from germinal center (GC) responses. However, it is unclear whether antiviral memory B cells represent a distinct subset among the highly heterogeneous memory B cell population. Here, we examined memory B cells induced by a virus-mimicking antigen at both transcriptome and epigenetic levels and found unexpectedly that antiviral memory B cells exhibit an enhanced innate immune response, which appeared to be facilitated by the epigenetic memory that is established through the memory B cell development. In addition, T-bet is associated with the altered chromatin architecture and is required for the formation of the antiviral memory B cells. Thus, antiviral memory B cells are distinct from other GC-derived memory B cells in both physiological functions and epigenetic landmarks.
Collapse
Affiliation(s)
- Xiping Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng Hong
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiachen Bu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yingping Liu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Can Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Runhan Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tiantian Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhuqiang Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Liping Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuyu Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaolin Hua
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Baidong Hou
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
34
|
Gadjalova I, Heinze JM, Goess MC, Hofmann J, Buck A, Weber MC, Blissenbach B, Kampick M, Krut O, Steiger K, Janssen KP, Neumann PA, Ruland J, Keppler SJ. B cell-mediated CD4 T-cell costimulation via CD86 exacerbates pro-inflammatory cytokine production during autoimmune intestinal inflammation. Mucosal Immunol 2024; 17:67-80. [PMID: 37918715 DOI: 10.1016/j.mucimm.2023.10.005] [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: 02/23/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/04/2023]
Abstract
Dysregulated B cell responses have been described in inflammatory bowel disease (IBD) patients; however, the role of B cells in IBD pathology remained incompletely understood. We here provide evidence for the detrimental role of activated B cells during the onset of autoimmune intestinal inflammation. Using Wiskott-Aldrich Syndrome interacting protein deficient (Wipf1-/-) mice as a mouse model of chronic colitis, we identified clusters of differentiation (CD)86 expression on activated B cells as a crucial factor exacerbating pro-inflammatory cytokine production of intestinal CD4 T cells. Depleting B cells through anti-CD20 antibody treatment or blocking costimulatory signals mediated by CD86 through cytotoxic T lymphocyte antigen-4-immunoglobulin (CTLA-4-Ig) diminished intestinal inflammation in our mouse model of chronic IBD at the onset of disease. This was due to a reduction in aberrant humoral immune responses and reduced CD4 T cell pro-inflammatory cytokine production, especially interferon-g (IFN-g) and granulocyte-macrophage colony-stimulating factor (GM-CSF). Interestingly, in addition to B cells isolated from the inflamed colon of Wipf1-/- mice, we also found CD86 mRNA and protein expression upregulated on activated B cells isolated from inflamed tissue of human patients with IBD. B cell activation and CD86 expression were boosted by soluble CD40L in vitro, which we found in the serum of mice and human patients with IBD. In summary, our data provides detailed insight into the contribution of B cells to intestinal inflammation, with implications for the treatment of IBD.
Collapse
Affiliation(s)
- Iana Gadjalova
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Julia M Heinze
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Marie C Goess
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Julian Hofmann
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Annalisa Buck
- Department of Surgery, Technical University of Munich, School of Medicine, Munich, Germany
| | - Marie-Christin Weber
- Department of Surgery, Technical University of Munich, School of Medicine, Munich, Germany
| | | | - Maximilian Kampick
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany
| | - Oleg Krut
- Paul-Ehrlich-Institut, Langen, Germany
| | - Katja Steiger
- Comparative Experimental Pathology, Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Technical University of Munich, School of Medicine, Munich, Germany
| | | | - Jürgen Ruland
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Center for Infection Research (DZIF), Munich, Germany
| | - Selina J Keppler
- Institute for Clinical Chemistry and Pathobiochemistry, Technical University of Munich, School of Medicine, Munich, Germany; TranslaTUM, Center for Translational Cancer Research, Technical University Munich, Munich, Germany; Division of Rheumatology and Clinical Immunology, Medical University Graz, Graz, Austria.
| |
Collapse
|
35
|
Najimi N, Kadi C, Elmtili N, Seghrouchni F, Bakri Y. Unravelling humoral immunity in SARS-CoV-2: Insights from infection and vaccination. Hum Antibodies 2024; 32:85-106. [PMID: 38758995 DOI: 10.3233/hab-230017] [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: 05/19/2024]
Abstract
Following infection and vaccination against SARS-CoV-2, humoral components of the adaptive immune system play a key role in protecting the host. Specifically, B cells generate high-affinity antibodies against various antigens of the virus. In this review, we discuss the mechanisms of immunity initiation through both natural infection and vaccination, shedding light on the activation of B cell subsets in response to SARS-CoV-2 infection and vaccination. The innate immune system serves as the initial line of primary and nonspecific defence against viruses. However, within several days following infection or a vaccine dose, a virus-specific immune response is initiated, primarily by B cells that produce antibodies. These antibodies contribute to the resolution of the disease. Subsequently, these B cells transition into memory B cells, which play a crucial role in providing long-term immunity against the virus. CD4+ T helper cells initiate a cascade, leading to B cell somatic hypermutation, germinal center memory B cells, and the production of neutralizing antibodies. B-cell dysfunction can worsen disease severity and reduce vaccine efficacy. Notably, individuals with B cell immunodeficiency show lower IL-6 production. Furthermore, this review delves into several aspects of immune responses, such as hybrid immunity, which has shown promise in boosting broad-spectrum protection. Cross-reactive immunity is under scrutiny as well, as pre-existing antibodies can offer protection against the disease. We also decipher breakthrough infection mechanisms, especially with the novel variants of the virus. Finally, we discuss some potential therapeutic solutions regarding B cells including convalescent plasma therapy, B-1 cells, B regulatory cell (Breg) modulation, and the use of neutralizing monoclonal antibodies in combating the infection. Ongoing research is crucial to grasp population immunity trends and assess the potential need for booster doses in maintaining effective immune responses against potential viral threats.
Collapse
Affiliation(s)
- Nouhaila Najimi
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Chaimae Kadi
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Noureddine Elmtili
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Fouad Seghrouchni
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Youssef Bakri
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| |
Collapse
|
36
|
Jiang Y, Chen H, Lin J, Pan J, Shen Y, Li Q. Anti-prolactin treatment alleviates lupus conditions by regulating the JAK2-STAT3 pathway. Clin Exp Pharmacol Physiol 2023; 50:936-943. [PMID: 37727880 DOI: 10.1111/1440-1681.13818] [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: 05/07/2023] [Revised: 08/12/2023] [Accepted: 08/16/2023] [Indexed: 09/21/2023]
Abstract
OBJECTIVES We previously revealed the role of prolactin (PRL) in antibody production and disease activity in patients with systemic lupus erythematosus. In this study, we sought to determine whether inhibition of PRL could improve lupus-like disease in MRL/lpr mice. METHODS The expression levels of PRL in various cell types of lupus patients were measured by flow cytometry. The effects of anti-PRL on animal survival, renal histopathology, creatinine, proteinuria, anti-dsDNA antibody, cytokine production, splenomegaly and lymphadenopathy were assessed. The effect of anti-PRL on the Jak2-Stat3 signalling pathway was detected by western blotting. RESULTS Prolactin was upregulated in B cells, neutrophils, CD4+ T cells, and monocytes isolated from patients with lupus. Furthermore, inhibition of PRL by anti-PRL treatment around the time of onset prolonged the survival of MRL/lpr mice, significantly reduced anti-dsDNA antibody production, and alleviated symptoms of lupus nephritis, splenomegaly, and lymphadenopathy. In addition, anti-PRL-treated mice showed a decrease in the levels of pathogenic cytokines such as IL-21 and IL-6. Furthermore, mechanistically, anti-PRL treatment significantly reduced the levels of p-Jak2 and p-Stat3 in MRL/lpr mice. CONCLUSIONS In summary, these data suggest that PRL inhibition alleviates lupus-like disease in MRL/lpr mice by modulating the Jak2-Stat3 signalling cascade. More importantly, our results imply the potential of PRL inhibitors and may provide a novel therapeutic approach for lupus.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Dermatology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Huyan Chen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, People's Republic of China
| | - Jinran Lin
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, People's Republic of China
| | - Jiewen Pan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, People's Republic of China
| | - Yanyun Shen
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, People's Republic of China
| | - Qiao Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, People's Republic of China
| |
Collapse
|
37
|
Hu S, Tao Y, Hu F, Liu X. Diminished LAG3 + B cells correlate with exacerbated rheumatoid arthritis. Ann Med 2023; 55:2208373. [PMID: 37143367 PMCID: PMC10165927 DOI: 10.1080/07853890.2023.2208373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Lymphocyte activation gene-3 (LAG3) positive B cells have been identified as a novel regulatory B cell subset, while the role of LAG3+ B cells in the pathogenesis of rheumatoid arthritis (RA) remains elusive. MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMCs) from RA, osteoarthritis (OA) patients and healthy volunteers were collected for flow cytometry staining of LAG3+ B cells. Their correlation with RA patient clinical and immunological features were analyzed. Moreover, the frequencies of LAG3+ B cells in collagen-induced arthritis (CIA) mice and naive mice were also detected. RESULTS A significant decrease of LAG3+ B cells was observed in RA patients as compared with healthy individuals and OA patients. Notably, the frequencies of LAG3+ B cells were negatively correlated with tender joint count (r = -0.4301, p = .0157) and DAS28-ESR (r = -0.4018, p = .025) in RA patients. In CIA mice, LAG3+ B cell frequencies were also decreased and negatively correlated with the CIA arthritis score. CONCLUSIONS Impairment of LAG3+ B cells potentially contributes to RA development. Reconstituting LAG3+ B cells might provide novel therapeutic strategies for the persistent disease.Key messagesLAG3+ B cells have been identified as a novel regulatory B cell subset. However, its role in the pathogenesis of RA remains unknown.This study revealed the decreased frequency of LAG3+ B cells in RA patients. Notably, LAG3+ B cells were negatively correlated with RA disease activity including the tender joint count and DAS28-ESR.In CIA mice, LAG3+ B cell frequencies were also decreased and negatively correlated with the CIA arthritis score.Reconstitution of LAG3+ B cells might provide novel therapeutic strategies for disease perpetuation.
Collapse
Affiliation(s)
- Suiyuan Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuting Tao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
38
|
Nepal D, Gazeley D. Role of IL-6 and IL-6 targeted therapy in systemic lupus erythematosus. Rheumatology (Oxford) 2023; 62:3804-3810. [PMID: 37594751 DOI: 10.1093/rheumatology/kead416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/13/2023] [Accepted: 07/16/2023] [Indexed: 08/19/2023] Open
Abstract
Interleukin-6 (IL-6) is one of the cytokines implicated in murine and human SLE. Only a few small studies have investigated IL-6 inhibition in human SLE. Currently, there are no studies registered in clinicaltrials.gov to assess the IL-6 targeted therapy in SLE, yet its role in the future remains to be defined. This narrative review analyses these and potential areas of future studies with IL-6 targeted therapy in SLE.
Collapse
Affiliation(s)
- Desh Nepal
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David Gazeley
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
39
|
Naithani U, Jain P, Sachan A, Khare P, Gabrani R. MicroRNA as a potential biomarker for systemic lupus erythematosus: pathogenesis and targeted therapy. Clin Exp Med 2023; 23:4065-4077. [PMID: 37921874 DOI: 10.1007/s10238-023-01234-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/24/2023] [Indexed: 11/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease associated with hyperactive innate and adaptive immune systems that cause dermatological, cardiovascular, renal, and neuropsychiatric problems in patients. SLE's multifactorial nature and complex pathogenesis present significant challenges in its clinical classification. In addition, unpredictable treatment responses in patients emphasize the need for highly specific and sensitive SLE biomarkers that can assist in understanding the exact pathogenesis and, thereby, lead to the identification of novel therapeutic targets. Recent studies on microRNA (miRNA), a non-coding region involved in the regulation of gene expression, indicate its importance in the development of the immune system and thus in the pathogenesis of various autoimmune disorders such as SLE. miRNAs are fascinating biomarker prospects for SLE categorization and disease monitoring owing to their small size and high stability. In this paper, we have discussed the involvement of a wide range of miRNAs in the regulation of SLE inflammation and how their modulation can be a potential therapeutic approach.
Collapse
Affiliation(s)
- Urshila Naithani
- Department of Biotechnology, A 10, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, 201309, India
| | - Priyanjal Jain
- Department of Biotechnology, A 10, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, 201309, India
| | - Aastha Sachan
- Department of Biotechnology, A 10, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, 201309, India
| | - Prachi Khare
- Department of Biotechnology, A 10, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, 201309, India
| | - Reema Gabrani
- Department of Biotechnology, A 10, Jaypee Institute of Information Technology, Sector-62, Noida, Uttar Pradesh, 201309, India.
| |
Collapse
|
40
|
Soni C, Makita S, Eichinger A, Serpas L, Sisirak V, Reizis B. Cutting Edge: TLR2 Signaling in B Cells Promotes Autoreactivity to DNA via IL-6 Secretion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1475-1480. [PMID: 37800687 PMCID: PMC10841863 DOI: 10.4049/jimmunol.2300313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
Autoantibodies to chromatin and dsDNA are a hallmark of systemic lupus erythematosus (SLE). In a mouse model of monogenic human SLE caused by DNASE1L3 deficiency, the anti-DNA response is dependent on endosomal nucleic acid-sensing TLRs TLR7 and TLR9. In this study, we report that this response also required TLR2, a surface receptor for microbial products that is primarily expressed on myeloid cells. Cell transfers into lymphopenic DNASE1L3-deficient mice showed that TLR2 was required for anti-DNA Ab production by lymphocytes. TLR2 was detectably expressed on B cells and facilitated the production of IL-6 by B cells activated in the presence of microbial products. Accordingly, treatment with broad-spectrum antibiotics or Ab-mediated blockade of IL-6 delayed the anti-DNA response in DNASE1L3-deficient mice. These studies reveal an unexpected B cell-intrinsic role of TLR2 in systemic autoreactivity to DNA, and they suggest that microbial products may synergize with self-DNA in the activation of autoreactive B cells in SLE.
Collapse
Affiliation(s)
- Chetna Soni
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sohei Makita
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anna Eichinger
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU, Munich, Germany
| | - Lee Serpas
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Vanja Sisirak
- CNRS-UMR 5164, ImmunoConcEpt, Université de Bordeaux, 33076 Bordeaux, France Dept. of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Boris Reizis
- Dept. of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
41
|
VURALLI D, DAĞIDIR HGÖK, TOPA EABBASOĞLU, BELEN HBOLAY. Leaky gut and inflammatory biomarkers in a medication overuse headache model in male rats. Turk J Med Sci 2023; 54:33-41. [PMID: 38812640 PMCID: PMC11031181 DOI: 10.55730/1300-0144.5763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/15/2024] [Accepted: 10/25/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Medication overuse is common among chronic migraine patients and nonsteroidal antiinflammatory drugs (NSAIDs) are the most frequently overused drugs. The pathophysiological mechanisms underlying medication overuse headache (MOH) are not completely understood. Intestinal hyperpermeability and leaky gut are reported in patients using NSAIDs. The aim of the study is to investigate the role of leaky gut and inflammation in an MOH model MOH model in male rats. Methods The study was conducted in male Sprague Dawley rats. There were two experimental groups. The first group was the chronic NSAID group in which the rats received mefenamic acid (n = 8) for four weeks intraperitoneally (ip) and the second group was the vehicle group (n = 8) that received 5% dimethyl sulfoxide+sesame oil (ip) for 4 weeks. We assessed spontaneous pain-like behavior, periorbital mechanical withdrawal thresholds, and anxiety-like behavior using an elevated plus maze test. After behavioral testing, serum levels of occludin and lipopolysaccharide-binding protein (LBP) and brain levels of IL-17, IL-6, and high mobility group box 1 protein (HMGB1) were evaluated with ELISA.Results: Serum LBP and occludin levels and brain IL-17 and HMGB1 levels were significantly elevated in the chronic NSAID group compared to its vehicle (p = 0.006, p = 0.016, p = 0.016 and p = 0.016 respectively) while brain IL-6 levels were comparable (p = 0.67) between the groups. The chronic NSAID group showed pain-like and anxiety-like behavior in behavioral tests. Brain IL-17 level was positively correlated with number of head shakes (r = 0.64, p = 0.045), brain IL-6 level was negatively correlated with periorbital mechanical withdrawal thresholds (r = -0.71, p = 0.049), and serum occludin level was positively correlated with grooming duration (r = 0.73, p = 0.032) in chronic NSAID group. Conclusion Elevated serum occludin and LBP levels and brain IL-17 and HMGB1 levels indicate a possible role of leaky gut and inflammation in an MOH model in male rats. Additionally, a significant correlation between pain behavior and markers of inflammation and intestinal hyperpermeability, supports the role of inflammation and leaky gut in MOH pathophysiology.
Collapse
Affiliation(s)
- Doğa VURALLI
- Department of Neurology and Algology, Faculty of Medicine, Gazi University, Ankara,
Turkiye
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Ankara,
Turkiye
- Neuropsychiatry Center, Gazi University, Ankara,
Turkiye
| | - Hale GÖK DAĞIDIR
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Ankara,
Turkiye
| | | | - Hayrunnisa BOLAY BELEN
- Department of Neurology and Algology, Faculty of Medicine, Gazi University, Ankara,
Turkiye
- Neuroscience and Neurotechnology Center of Excellence (NÖROM), Gazi University, Ankara,
Turkiye
- Neuropsychiatry Center, Gazi University, Ankara,
Turkiye
| |
Collapse
|
42
|
Wilson JJ, Wei J, Daamen AR, Sears JD, Bechtel E, Mayberry CL, Stafford GA, Bechtold L, Grammer AC, Lipsky PE, Roopenian DC, Chang CH. Glucose oxidation-dependent survival of activated B cells provides a putative novel therapeutic target for lupus treatment. iScience 2023; 26:107487. [PMID: 37636066 PMCID: PMC10448027 DOI: 10.1016/j.isci.2023.107487] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/27/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Aberrant metabolic demand is observed in immune/inflammatory disorders, yet the role in pathogenesis remains unclear. Here, we discover that in lupus, activated B cells, including germinal center B (GCB) cells, have remarkably high glycolytic requirement for survival over T cell populations, as demonstrated by increased metabolic activity in lupus-activated B cells compared to immunization-induced cells. The augmented reliance on glucose oxidation makes GCB cells vulnerable to mitochondrial ROS-induced oxidative stress and apoptosis. Short-term glycolysis inhibition selectively reduces pathogenic activated B in lupus-prone mice, extending their lifespan, without affecting T follicular helper cells. Particularly, BCMA-expressing GCB cells rely heavily on glucose oxidation. Depleting BCMA-expressing activated B cells with APRIL-based CAR-T cells significantly prolongs the lifespan of mice with severe autoimmune disease. These results reveal that glycolysis-dependent activated B and GCB cells, especially those expressing BCMA, are potentially key lupus mediators, and could be targeted to improve disease outcomes.
Collapse
Affiliation(s)
- John J. Wilson
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | - Jian Wei
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Andrea R. Daamen
- AMPEL BioSolutions and the RILITE Research Institute, Charlottesville, VA 22902, USA
| | - John D. Sears
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elaine Bechtel
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
| | | | | | | | - Amrie C. Grammer
- AMPEL BioSolutions and the RILITE Research Institute, Charlottesville, VA 22902, USA
| | - Peter E. Lipsky
- AMPEL BioSolutions and the RILITE Research Institute, Charlottesville, VA 22902, USA
| | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, Maine, ME 04609, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
43
|
Li Y, Zhu L, Ko CJ, Yang JY, Wang H, Manyam G, Wang J, Cheng X, Zhao S, Jie Z. TRAF3-EWSR1 signaling axis acts as a checkpoint on germinal center responses. J Exp Med 2023; 220:e20221483. [PMID: 37097293 PMCID: PMC10130905 DOI: 10.1084/jem.20221483] [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: 08/29/2022] [Revised: 12/29/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
The formation of germinal centers (GCs) is crucial for humoral immunity and vaccine efficacy. Constant stimulation through microbiota drives the formation of constitutive GCs in Peyer's patches (PPs), which generate B cells that produce antibodies against gut antigens derived from commensal bacteria and infectious pathogens. However, the molecular mechanism that regulates this persistent process is poorly understood. We report that Ewing Sarcoma Breakpoint Region 1 (EWSR1) is a brake to constitutive GC generation and immunoglobulin G (IgG) production in PPs, vaccination-induced GC formation, and IgG responses. Mechanistically, EWSR1 suppresses Bcl6 upregulation after antigen encounter, thereby negatively regulating induced GC B cell generation and IgG production. We further showed that tumor necrosis factor receptor-associated factor (TRAF) 3 serves as a negative regulator of EWSR1. These results established that the TRAF3-EWSR1 signaling axis acts as a checkpoint for Bcl6 expression and GC responses, indicating that this axis is a therapeutic target to tune GC responses and humoral immunity in infectious diseases.
Collapse
Affiliation(s)
- Yanchuan Li
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Lele Zhu
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA
| | - Chun-Jung Ko
- Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jin-Young Yang
- Department of Biological Sciences, Pusan National University, Busan, Korea
| | - Hongjiao Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Ganiraju Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuhong Cheng
- Memorial Hermann-Texas Medical Center, Houston, TX, USA
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Zuliang Jie
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
44
|
Xuan T, Yuan X, Zheng S, Wang L, Wang Q, Zhang S, Qi F, Luan W. Repeated Lipoteichoic Acid Injection at Low Concentration Induces Capsular Contracture by Activating Adaptive Immune Response through the IL-6/STAT3 Signaling Pathway. Plast Reconstr Surg 2023; 152:349-359. [PMID: 36700876 DOI: 10.1097/prs.0000000000010224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Capsular contracture is the most common complication of breast implantation surgery. Bacterial contamination was considered to play an important role in the occurrence of capsular contracture, and Gram-positive bacteria such as Staphylococcus epidermidis were discovered in the clinical specimens. Lipoteichoic acid (LTA) was a component of the cell wall of Gram-positive bacteria and was sufficient in the pathogenicity of the bacteria. The authors assumed that LTA could trigger the immunologic response against the implant and cause capsular contracture. METHODS The authors developed a rat model of capsular contracture by repeated injection of 10 μg/mL LTA. The histologic changes of the capsule tissue were measured by hematoxylin and eosin, sirius red, Masson, and immunohistochemical staining. The expression of related cytokines was measured by quantitative real-time polymerase chain reaction. The downstream pathway activation was shown by Western blot. The authors also applied tocilizumab, an interleukin (IL)-6 receptor antagonist, to verify the role of IL-6 in this pathologic process. RESULTS The authors discovered that repeated LTA injection, at a low concentration, could induce the thickening of capsule tissue, the deposition of collagen fiber, and the activation of myofibroblasts. The IL-6/signal transducer and activator of transcription 3 signaling pathway was activated in this process, and the inhibition of IL-6 receptor could relieve the symptoms. B cells and T-helper cells, especially T-helper type 1, could be related to this phenomenon. CONCLUSIONS The authors' research corroborated that subclinical infection could trigger capsular contracture, and the immune system played an important role in this process. The authors' results provided a possible research direction for the mechanism of bacterial infection-induced immune response against breast implants. CLINICAL RELEVANCE STATEMENT The authors' research provides a possible research direction for the mechanism of bacterial infection-induced immune response against breast implants, and a potential target for predicting the prognosis of capsular contracture.
Collapse
Affiliation(s)
- Tianfan Xuan
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University
- Treatment Center of Burn and Trauma, Affiliated Hospital of Jiangnan University, Jiangnan University
| | - Xin Yuan
- Department of Plastic and Burn Surgery, West China School of Medicine, West China Hospital, Sichuan University
| | - Shaoluan Zheng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University (Xiamen Branch)
| | - Lu Wang
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University
| | - Qiang Wang
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University
| | - Simin Zhang
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University
| | - Fazhi Qi
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University
| | - Wenjie Luan
- From the Department of Plastic Surgery, Zhongshan Hospital, Fudan University
| |
Collapse
|
45
|
Chunder R, Schropp V, Marzin M, Amor S, Kuerten S. A Dual Role of Osteopontin in Modifying B Cell Responses. Biomedicines 2023; 11:1969. [PMID: 37509608 PMCID: PMC10377065 DOI: 10.3390/biomedicines11071969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The occurrence of B cell aggregates within the central nervous system (CNS) has prompted the investigation of the potential sources of pathogenic B cell and T cell responses in a subgroup of secondary progressive multiple sclerosis (MS) patients. Nevertheless, the expression profile of molecules associated with these aggregates and their role in aggregate development and persistence is poorly described. Here, we focused on the expression pattern of osteopontin (OPN), which is a well-described cytokine, in MS brain tissue. Autopsied brain sections from MS cases with and without B cell pathology were screened for the presence of CD20+ B cell aggregates and co-expression of OPN. To demonstrate the effect of OPN on B cells, flow cytometry, ELISA and in vitro aggregation assays were conducted using the peripheral blood of healthy volunteers. Although OPN was expressed in MS brain tissue independent of B cell pathology, it was also highly expressed within B cell aggregates. In vitro studies demonstrated that OPN downregulated the co-stimulatory molecules CD80 and CD86 on B cells. OPN-treated B cells produced significantly lower amounts of IL-6. However, OPN-treated B cells also exhibited a higher tendency to form homotypic cell aggregates in vitro. Taken together, our data indicate a conflicting role of OPN in modulating B cell responses.
Collapse
Affiliation(s)
- Rittika Chunder
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Verena Schropp
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Manuel Marzin
- Department of Pathology, Amsterdam University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam University Medical Center, 1007 MB Amsterdam, The Netherlands
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Stefanie Kuerten
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
46
|
Largent AD, Lambert K, Chiang K, Shumlak N, Liggitt D, Oukka M, Torgerson TR, Buckner JH, Allenspach EJ, Rawlings DJ, Jackson SW. Dysregulated IFN-γ signals promote autoimmunity in STAT1 gain-of-function syndrome. Sci Transl Med 2023; 15:eade7028. [PMID: 37406138 PMCID: PMC11645977 DOI: 10.1126/scitranslmed.ade7028] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 06/16/2023] [Indexed: 07/07/2023]
Abstract
Heterozygous signal transducer and activator of transcription 1 (STAT1) gain-of-function (GOF) mutations promote a clinical syndrome of immune dysregulation characterized by recurrent infections and predisposition to humoral autoimmunity. To gain insights into immune characteristics of STAT1-driven inflammation, we performed deep immunophenotyping of pediatric patients with STAT1 GOF syndrome and age-matched controls. Affected individuals exhibited dysregulated CD4+ T cell and B cell activation, including expansion of TH1-skewed CXCR3+ populations that correlated with serum autoantibody titers. To dissect underlying immune mechanisms, we generated Stat1 GOF transgenic mice (Stat1GOF mice) and confirmed the development of spontaneous humoral autoimmunity that recapitulated the human phenotype. Despite clinical resemblance to human regulatory T cell (Treg) deficiency, Stat1GOF mice and humans with STAT1 GOF syndrome exhibited normal Treg development and function. In contrast, STAT1 GOF autoimmunity was characterized by adaptive immune activation driven by dysregulated STAT1-dependent signals downstream of the type 1 and type 2 interferon (IFN) receptors. However, in contrast to the prevailing type 1 IFN-centric model for STAT1 GOF autoimmunity, Stat1GOF mice lacking the type 1 IFN receptor were only partially protected from STAT1-driven systemic inflammation, whereas loss of type 2 IFN (IFN-γ) signals abrogated autoimmunity. Last, germline STAT1 GOF alleles are thought to enhance transcriptional activity by increasing total STAT1 protein, but the underlying biochemical mechanisms have not been defined. We showed that IFN-γ receptor deletion normalized total STAT1 expression across immune lineages, highlighting IFN-γ as the critical driver of feedforward STAT1 elevation in STAT1 GOF syndrome.
Collapse
Affiliation(s)
| | | | - Kristy Chiang
- Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Natali Shumlak
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Present address: Division of Medical Genetics, University of Washington School of Medicine; Seattle, WA 98195, USA
| | - Denny Liggitt
- Department of Comparative Medicine, University of Washington School of Medicine; Seattle, WA 98195, USA
| | - Mohammed Oukka
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA 98195, USA
- Department of Immunology, University of Washington School of Medicine; Seattle, WA 98195, USA
| | | | | | - Eric J. Allenspach
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA 98195, USA
| | - David J. Rawlings
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA 98195, USA
- Department of Immunology, University of Washington School of Medicine; Seattle, WA 98195, USA
| | - Shaun W. Jackson
- Seattle Children's Research Institute, Seattle, WA 98101, USA
- Department of Pediatrics, University of Washington School of Medicine; Seattle, WA 98195, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine; Seattle, WA 98195, USA
| |
Collapse
|
47
|
Mantel I, Fein MR, Donlin LT. Emerging synovial cell states in rheumatoid arthritis as potential therapeutic targets. Curr Opin Rheumatol 2023; 35:249-254. [PMID: 37040654 PMCID: PMC10219846 DOI: 10.1097/bor.0000000000000940] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
PURPOSE OF REVIEW To summarize recently discovered novel cell states in rheumatoid arthritis (RA) synovium that could have important implications for disease treatment. RECENT FINDINGS The use of multiomic technologies, including single-cell and spatial transcriptomics and mass cytometry, has led to the discovery of several novel cell states, which could have important implications for the treatment of RA. These cells can be found in patient blood, synovial fluid, or synovial tissue and span several immune cell subsets as well as stromal cell types. These diverse cell states may represent the targets of current or future therapeutics, while their fluctuations may inform the ideal timing for therapy. Future efforts are needed to implicate how each cell state functions in the pathophysiologic network within affected joints and how medications perturb each cell state and ultimately the tissue. SUMMARY Multiomic molecular technologies have afforded the discovery of numerous novel cellular states in RA synovium; the next challenge will be to link these states to pathophysiology and treatment response.
Collapse
Affiliation(s)
- Ian Mantel
- Weill Cornell Medicine Graduate School
- Hospital for Special Surgery Research Institute, New York, New York, USA
| | - Miriam R Fein
- Hospital for Special Surgery Research Institute, New York, New York, USA
| | - Laura T Donlin
- Weill Cornell Medicine Graduate School
- Hospital for Special Surgery Research Institute, New York, New York, USA
| |
Collapse
|
48
|
Camici M, Garcia-Gil M, Allegrini S, Pesi R, Bernardini G, Micheli V, Tozzi MG. Inborn Errors of Purine Salvage and Catabolism. Metabolites 2023; 13:787. [PMID: 37512494 PMCID: PMC10383617 DOI: 10.3390/metabo13070787] [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: 04/27/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Cellular purine nucleotides derive mainly from de novo synthesis or nucleic acid turnover and, only marginally, from dietary intake. They are subjected to catabolism, eventually forming uric acid in humans, while bases and nucleosides may be converted back to nucleotides through the salvage pathways. Inborn errors of the purine salvage pathway and catabolism have been described by several researchers and are usually referred to as rare diseases. Since purine compounds play a fundamental role, it is not surprising that their dysmetabolism is accompanied by devastating symptoms. Nevertheless, some of these manifestations are unexpected and, so far, have no explanation or therapy. Herein, we describe several known inborn errors of purine metabolism, highlighting their unexplained pathological aspects. Our intent is to offer new points of view on this topic and suggest diagnostic tools that may possibly indicate to clinicians that the inborn errors of purine metabolism may not be very rare diseases after all.
Collapse
Affiliation(s)
- Marcella Camici
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Mercedes Garcia-Gil
- Unità di Fisiologia Generale, Dipartimento di Biologia, Università di Pisa, Via San Zeno 31, 56127 Pisa, Italy
- CISUP, Centro per l'Integrazione Della Strumentazione Dell'Università di Pisa, 56127 Pisa, Italy
- Centro di Ricerca Interdipartimentale Nutrafood "Nutraceuticals and Food for Health", Università di Pisa, 56126 Pisa, Italy
| | - Simone Allegrini
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
- CISUP, Centro per l'Integrazione Della Strumentazione Dell'Università di Pisa, 56127 Pisa, Italy
- Centro di Ricerca Interdipartimentale Nutrafood "Nutraceuticals and Food for Health", Università di Pisa, 56126 Pisa, Italy
| | - Rossana Pesi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| | - Giulia Bernardini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Vanna Micheli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
- LND Famiglie Italiane ODV-Via Giovanetti 15-20, 16149 Genova, Italy
| | - Maria Grazia Tozzi
- Unità di Biochimica, Dipartimento di Biologia, Università di Pisa, Via San Zeno 51, 56127 Pisa, Italy
| |
Collapse
|
49
|
Gunnarsdottir FB, Briem O, Lindgren AY, Källberg E, Andersen C, Grenthe R, Rosenqvist C, Millrud CR, Wallgren M, Viklund H, Bexell D, Johansson ME, Hedenfalk I, Hagerling C, Leandersson K. Breast cancer associated CD169 + macrophages possess broad immunosuppressive functions but enhance antibody secretion by activated B cells. Front Immunol 2023; 14:1180209. [PMID: 37404831 PMCID: PMC10315498 DOI: 10.3389/fimmu.2023.1180209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
CD169+ resident macrophages in lymph nodes of breast cancer patients are for unknown reasons associated with a beneficial prognosis. This contrasts CD169+ macrophages present in primary breast tumors (CD169+ TAMs), that correlate with a worse prognosis. We recently showed that these CD169+ TAMs were associated with tertiary lymphoid structures (TLSs) and Tregs in breast cancer. Here, we show that CD169+ TAMs can be monocyte-derived and express a unique mediator profile characterized by type I IFNs, CXCL10, PGE2 and inhibitory co-receptor expression pattern. The CD169+ monocyte-derived macrophages (CD169+ Mo-M) possessed an immunosuppressive function in vitro inhibiting NK, T and B cell proliferation, but enhanced antibody and IL6 secretion in activated B cells. Our findings indicate that CD169+ Mo-M in the primary breast tumor microenvironment are linked to both immunosuppression and TLS functions, with implications for future targeted Mo-M therapy.
Collapse
Affiliation(s)
- Frida Björk Gunnarsdottir
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Oscar Briem
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Aida Yifter Lindgren
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Eva Källberg
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Cajsa Andersen
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Robert Grenthe
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Cassandra Rosenqvist
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Camilla Rydberg Millrud
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Mika Wallgren
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Hannah Viklund
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| | - Daniel Bexell
- Translational Cancer Research, TCR, Medicon Village, Lund University, Lund, Sweden
| | - Martin E. Johansson
- Sahlgrenska Center for Cancer Research, Department of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Catharina Hagerling
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
- Division of Clinical Genetics, Department of Laboratory Medicine Lund, Lund University, Lund, Sweden
| | - Karin Leandersson
- Cancer Immunology, Department for Translational Medicine, Clinical Research Center, Lund University, Malmö, Sweden
| |
Collapse
|
50
|
Bachus H, McLaughlin E, Lewis C, Papillion AM, Benveniste EN, Hill DD, Rosenberg AF, Ballesteros-Tato A, León B. IL-6 prevents Th2 cell polarization by promoting SOCS3-dependent suppression of IL-2 signaling. Cell Mol Immunol 2023; 20:651-665. [PMID: 37046042 PMCID: PMC10229632 DOI: 10.1038/s41423-023-01012-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
Defective interleukin-6 (IL-6) signaling has been associated with Th2 bias and elevated IgE levels. However, the underlying mechanism by which IL-6 prevents the development of Th2-driven diseases remains unknown. Using a model of house dust mite (HDM)-induced Th2 cell differentiation and allergic airway inflammation, we showed that IL-6 signaling in allergen-specific T cells was required to prevent Th2 cell differentiation and the subsequent IgE response and allergic inflammation. Th2 cell lineage commitment required strong sustained IL-2 signaling. We found that IL-6 turned off IL-2 signaling during early T-cell activation and thus inhibited Th2 priming. Mechanistically, IL-6-driven inhibition of IL-2 signaling in responding T cells was mediated by upregulation of Suppressor Of Cytokine Signaling 3 (SOCS3). This mechanism could be mimicked by pharmacological Janus Kinase-1 (JAK1) inhibition. Collectively, our results identify an unrecognized mechanism that prevents the development of unwanted Th2 cell responses and associated diseases and outline potential preventive interventions.
Collapse
Affiliation(s)
- Holly Bachus
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erin McLaughlin
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Crystal Lewis
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amber M Papillion
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Alexion Pharmaceuticals, Inc., New Haven, CT, USA
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dave Durell Hill
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexander F Rosenberg
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Informatics Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - André Ballesteros-Tato
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Beatriz León
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|