1
|
Steffen TL, Stafford JD, Samson WK, Yosten GLC. Nesfatin-1 is a regulator of inflammation with implications during obesity and metabolic syndrome. Appetite 2024; 203:107669. [PMID: 39251090 DOI: 10.1016/j.appet.2024.107669] [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: 02/01/2024] [Revised: 08/19/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024]
Abstract
Nesfatin-1, derived from the nucleobindin 2 (NUCB2) precursor, is a potent anorexigenic peptide that was discovered in 2006. Since its identification in the hypothalamus, it has been shown to have wide ranging actions within and outside of the central nervous system. One of these actions is the regulation of inflammation, which could potentially be exploited therapeutically in the context of obesity-associated inflammation in adipose tissue. Here, we review recent advances in our knowledge about the ability of nesfatin-1 to control inflammation by regulating NFκB signaling, which likely attenuates pro-inflammatory cytokine production and inhibits apoptosis.
Collapse
Affiliation(s)
- Tara L Steffen
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, St. Louis, MO, USA.
| | - Joshua D Stafford
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, St. Louis, MO, USA
| | - Willis K Samson
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, St. Louis, MO, USA
| | - Gina L C Yosten
- Saint Louis University School of Medicine, Department of Pharmacology and Physiology, St. Louis, MO, USA
| |
Collapse
|
2
|
Ge Y, Liu H, Huang W, Zhu H, Zong D, He X. Immunoinhibitory effects of hypoxia-driven reprogramming of EGR1 hi and EGR3 positive B cells in the nasopharyngeal carcinoma microenvironment. Oral Oncol 2024; 158:106999. [PMID: 39197193 DOI: 10.1016/j.oraloncology.2024.106999] [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: 06/29/2024] [Revised: 07/27/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024]
Abstract
Regulatory B (Breg) cells is a type of immune cell that exhibit immunosuppressive behavior within the tumor microenvironment. However, the differentiation and regulatory mechanisms of these Breg cells remain unexplored. Single-cell transcriptome sequencing analysis of human nasopharyngeal carcinoma (NPC) revealed a significant enrichment of B cell subset characterized by high expression of EGR1 and EGR3 in the tumor microenvironment. Notably, in the hypoxic microenvironment, these B cells induce MAPK pathway activation, subsequently triggering the activation of transcription factors EGR1 and EGR3, which further modulate the expression of immunosuppressive factors like TGFB1 and IL10. In transplant experiments using primary B cells induced under hypoxia and co-transplanted with cancer cells, a significant increase in tumor growth was observed. Mechanism experiments demonstrated that EGR1hi and EGR3+ B cells further activate the maturation and immunosuppressive function of Treg cells through the secretion of IL16 and TNF-α. Hence, this study identifies the key transcription factors EGR1 and EGR3 as essential regulators and elucidates the differentiation of Breg cells under hypoxic conditions.
Collapse
Affiliation(s)
- Yizhi Ge
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Haitao Liu
- College of Life Science, Inner Mongolia University, Hohhot 010021, China
| | - Wenxuan Huang
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Huanfeng Zhu
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Dan Zong
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China; Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Bradford HF, Mauri C. Diversity of regulatory B cells: Markers and functions. Eur J Immunol 2024; 54:e2350496. [PMID: 39086053 DOI: 10.1002/eji.202350496] [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: 03/20/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Regulatory B cells (Bregs) are a functionally distinct B-cell subset involved in the maintenance of homeostasis and inhibition of inflammation. Studies, from the last two decades, have increased our understanding of cellular and molecular mechanisms involved in their generation, function, and to a certain extent phenotype. Current research endeavours to unravel the causes and consequences of Breg defects in disease, with increasing evidence highlighting the relevance of Bregs in promoting tumorigenic responses. Here we provide historical and emerging findings of the significance of Bregs in autoimmunity and transplantation, and how these insights have translated into the cancer field.
Collapse
Affiliation(s)
- Hannah F Bradford
- Division of Infection and Immunity and Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| | - Claudia Mauri
- Division of Infection and Immunity and Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, United Kingdom
| |
Collapse
|
4
|
Puppa MA, Bennstein SB, Fischer HJ, Rink L. Zinc deficiency impairs the development of human regulatory B cells from purified B cells. J Trace Elem Med Biol 2024; 86:127556. [PMID: 39442468 DOI: 10.1016/j.jtemb.2024.127556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Zinc is a vital trace element, important for many different immune processes and adequate functionality. B cell development is known to be dependent on sufficient zinc supply. Recently a regulatory B cell (Breg) population has been identified, as CD19+IL-10+ B cells, able to regulate immune responses by secretion of anti-inflammatory cytokines, such as IL-10. Due to their promotion of an anti-inflammatory milieu, Bregs could reduce or might even prevent excessive pro-inflammatory responses. Hence, having and maintaining Bregs could be interesting for patients suffering from allergies, asthma, and autoimmune diseases. Therefore, understanding Breg generation, required signaling, and their developmental requirements are important. Since our group could previously show that zinc is important for regulatory T cells, we aimed to determine the effect of zinc deficiency on Breg development from human peripheral blood CD19+ B cells. We observed highest Breg generation with a combined stimulus of CD40L and the toll like receptor (TLR) ligand, CpG-ODN2006. Using this stimulus, we observed that zinc deficient medium significantly decreased Breg generation from purified B cells. This was not seen in Bregs generated from peripheral blood mononuclear cells (PBMCs) without B cell enrichment suggesting a compensatory mechanism. In line with literature, our data also confirms Bregs develop from CD19+ B cells, since total CD19+ frequencies remained unchanged, while Breg frequencies varied between stimuli and zinc media conditions. Our study shows for the first time that zinc deficiency significantly impairs Breg development, which provides an important new perspective for clinical applications and therapeutic strategies.
Collapse
Affiliation(s)
- Mary-Ann Puppa
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Sabrina B Bennstein
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Henrike J Fischer
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Lothar Rink
- Institute of Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
5
|
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] [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
|
6
|
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
|
7
|
Liang Y, Li Y, Lee C, Yu Z, Chen C, Liang C. Ulcerative colitis: molecular insights and intervention therapy. MOLECULAR BIOMEDICINE 2024; 5:42. [PMID: 39384730 PMCID: PMC11464740 DOI: 10.1186/s43556-024-00207-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by abdominal pain, diarrhea, rectal bleeding, and weight loss. The pathogenesis and treatment of UC remain key areas of research interest. Various factors, including genetic predisposition, immune dysregulation, and alterations in the gut microbiota, are believed to contribute to the pathogenesis of UC. Current treatments for UC include 5-aminosalicylic acids, corticosteroids, immunosuppressants, and biologics. However, study reported that the one-year clinical remission rate is only around 40%. It is necessary to prompt the exploration of new treatment modalities. Biologic therapies, such as anti-TNF-α monoclonal antibody and JAK inhibitor, primarily consist of small molecules targeting specific pathways, effectively inducing and maintaining remission. Given the significant role of the gut microbiota, research into intestinal microecologics, such as probiotics and prebiotics, and fecal microbiota transplantation (FMT) shows promising potential in UC treatment. Additionally, medicinal herbs, such as chili pepper and turmeric, used in complementary therapy have shown promising results in UC management. This article reviews recent findings on the mechanisms of UC, including genetic susceptibility, immune cell dynamics and cytokine regulation, and gut microbiota alterations. It also discusses current applications of biologic therapy, herbal therapy, microecologics, and FMT, along with their prospects and challenges.
Collapse
Affiliation(s)
- Yuqing Liang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yang Li
- Department of Respiratory, Sichuan Integrative Medicine Hospital, Chengdu, 610042, China
| | - Chehao Lee
- Department of Traditional Chinese Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ziwei Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Chongli Chen
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Chao Liang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
8
|
Baert L, Mahmudul HM, Stegall M, Joo H, Oh S. B Cell-mediated Immune Regulation and the Quest for Transplantation Tolerance. Transplantation 2024; 108:2021-2033. [PMID: 38389135 DOI: 10.1097/tp.0000000000004948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Pathophysiologic function of B cells in graft rejection has been well recognized in transplantation. B cells promote alloantigen-specific T-cell response and secrete antibodies that can cause antibody-mediated graft failures and rejections. Therefore, strategies targeting B cells, for example, B-cell depletion, have been used for the prevention of both acute and chronic rejections. Interestingly, however, recent mounting evidence indicates that subsets of B cells yet to be further identified can display potent immune regulatory functions, and they contribute to transplantation tolerance and operational tolerance in both experimental and clinical settings, respectively. In this review, we integrate currently available information on B-cell subsets, including T-cell Ig domain and mucin domain 1-positive transitional and T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif domain-positive memory B cells, displaying immune regulatory functions, with a focus on transplantation tolerance, by analyzing their mechanisms of action. In addition, we will discuss potential T-cell Ig domain and mucin domain 1-positive and T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif domain-positive B cell-based strategies for the enhancement of operational tolerance in transplantation patients.
Collapse
Affiliation(s)
- Laurie Baert
- Department of Immunology, Mayo Clinic, Scottsdale, AZ
| | | | - Mark Stegall
- Department of Surgery, William J. von Liebig Transplant Center, Mayo Clinic, Rochester, MN
| | - HyeMee Joo
- Department of Immunology, Mayo Clinic, Scottsdale, AZ
| | - SangKon Oh
- Department of Immunology, Mayo Clinic, Scottsdale, AZ
| |
Collapse
|
9
|
Ahsan NF, Lourenço S, Psyllou D, Long A, Shankar S, Bashford-Rogers R. The current understanding of the phenotypic and functional properties of human regulatory B cells (Bregs). OXFORD OPEN IMMUNOLOGY 2024; 5:iqae012. [PMID: 39346706 PMCID: PMC11427547 DOI: 10.1093/oxfimm/iqae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/13/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024] Open
Abstract
B cells can have a wide range of pro- and anti- inflammatory functions. A subset of B cells called regulatory B cells (Bregs) can potently suppress immune responses. Bregs have been shown to maintain immune homeostasis and modulate inflammatory responses. Bregs are an exciting cellular target across a range of diseases, including Breg induction in autoimmunity, allergy and transplantation, and Breg suppression in cancers and infection. Bregs exhibit a remarkable phenotypic heterogeneity, rendering their unequivocal identification a challenging task. The lack of a universally accepted and exclusive surface marker set for Bregs across various studies contributes to inconsistencies in their categorization. This review paper presents a comprehensive overview of the current understanding of the phenotypic and functional properties of human Bregs while addressing the persisting ambiguities and discrepancies in their characterization. Finally, the paper examines the promising therapeutic opportunities presented by Bregs as their immunomodulatory capacities have gained attention in the context of autoimmune diseases, allergic conditions, and cancer. We explore the exciting potential in harnessing Bregs as potential therapeutic agents and the avenues that remain open for the development of Breg-based treatment strategies.
Collapse
Affiliation(s)
- Nawara Faiza Ahsan
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
- Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Stella Lourenço
- Keizo Asami Institute, Federal University of Pernambuco, Recife 50740-520, Brazil
| | - Dimitra Psyllou
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Alexander Long
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
| | - Sushma Shankar
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - Rachael Bashford-Rogers
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, United Kingdom
- Oxford Cancer Centre, University of Oxford, Oxford OX3 7LH, United Kingdom
| |
Collapse
|
10
|
Martínez LE, Comin-Anduix B, Güemes-Aragon M, Ibarrondo J, Detels R, Mimiaga MJ, Epeldegui M. Characterization of unique B-cell populations in the circulation of people living with HIV prior to non-Hodgkin lymphoma diagnosis. Front Immunol 2024; 15:1441994. [PMID: 39324141 PMCID: PMC11422120 DOI: 10.3389/fimmu.2024.1441994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/20/2024] [Indexed: 09/27/2024] Open
Abstract
People living with HIV (PLWH) are at higher risk of developing lymphoma. In this study, we performed cytometry by time-of-flight (CyTOF) on peripheral blood mononuclear cells of cART-naïve HIV+ individuals and cART-naïve HIV+ individuals prior to AIDS-associated non-Hodgkin lymphoma (pre-NHL) diagnosis. Participants were enrolled in the Los Angeles site of the MACS/WIHS Combined Cohort Study (MWCCS). Uniform Manifold Approximation and Projection (UMAP) and unsupervised clustering analysis were performed to identify differences in the expression of B-cell activation markers and/or oncogenic markers associated with lymphomagenesis. CD10+CD27- B cells, CD20+CD27- B cells, and B-cell populations with aberrant features (CD20+CD27+CXCR4+CD71+ B cells and CD20+CXCR4+cMYC+ B cells) were significantly elevated in HIV+ cART-naïve compared to HIV-negative samples. CD20+CD27+CD24+CXCR4+CXCR5+ B cells, CD20+CD27+CD10+CD24+CXCR4+cMYC+ B cells, and a cluster of CD20+CXCR4hiCD27-CD24+CXCR5+CD40+CD4+AICDA+ B cells were significantly elevated in HIV+ pre-NHL (cART-naïve) compared to HIV+ cART-naïve samples. A potentially clonal cluster of CD20+CXCR4+CXCR5+cMYC+AICDA+ B cells and a cluster of germinal center B-cell-like cells (CD19-CD20+CXCR4+Bcl-6+PD-L1+cMYC+) were also found in the circulation of HIV+ pre-NHL (cART-naïve) samples. Moreover, significantly elevated clusters of CD19+CD24hiCD38hi cMYC+ AICDA+ B regulatory cells were identified in HIV+ pre-NHL (cART-naïve) compared to HIV+ cART-naïve samples. The present study identifies unique B-cell subsets in PLWH with potential pre-malignant features that may contribute to the development of pre-tumor B cells in PLWH and that may play a role in lymphomagenesis.
Collapse
Affiliation(s)
- Laura E Martínez
- UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Begoña Comin-Anduix
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Surgical Oncology, Department of Surgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Miriam Güemes-Aragon
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Javier Ibarrondo
- UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Roger Detels
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
| | - Matthew J Mimiaga
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA, United States
| | - Marta Epeldegui
- UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
Yang Y, Chen X, Pan J, Ning H, Zhang Y, Bo Y, Ren X, Li J, Qin S, Wang D, Chen MM, Zhang Z. Pan-cancer single-cell dissection reveals phenotypically distinct B cell subtypes. Cell 2024; 187:4790-4811.e22. [PMID: 39047727 DOI: 10.1016/j.cell.2024.06.038] [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: 06/09/2023] [Revised: 04/25/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Characterizing the compositional and phenotypic characteristics of tumor-infiltrating B cells (TIBs) is important for advancing our understanding of their role in cancer development. Here, we establish a comprehensive resource of human B cells by integrating single-cell RNA sequencing data of B cells from 649 patients across 19 major cancer types. We demonstrate substantial heterogeneity in their total abundance and subtype composition and observe immunoglobulin G (IgG)-skewness of antibody-secreting cell isotypes. Moreover, we identify stress-response memory B cells and tumor-associated atypical B cells (TAABs), two tumor-enriched subpopulations with prognostic potential, shared in a pan-cancer manner. In particular, TAABs, characterized by a high clonal expansion level and proliferative capacity as well as by close interactions with activated CD4 T cells in tumors, are predictive of immunotherapy response. Our integrative resource depicts distinct clinically relevant TIB subsets, laying a foundation for further exploration of functional commonality and diversity of B cells in cancer.
Collapse
Affiliation(s)
- Yu Yang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xueyan Chen
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jieying Pan
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Huiheng Ning
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Department of Liver Surgery, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yufei Bo
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Xianwen Ren
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiesheng Li
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Shishang Qin
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China
| | - Dongfang Wang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| | - Min-Min Chen
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518132, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, and School of Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
12
|
Gilio L, Fresegna D, Stampanoni Bassi M, Musella A, De Vito F, Balletta S, Sanna K, Caioli S, Pavone L, Galifi G, Simonelli I, Guadalupi L, Vanni V, Buttari F, Dolcetti E, Bruno A, Azzolini F, Borrelli A, Fantozzi R, Finardi A, Furlan R, Centonze D, Mandolesi G. Interleukin-10 contrasts inflammatory synaptopathy and central neurodegenerative damage in multiple sclerosis. Front Mol Neurosci 2024; 17:1430080. [PMID: 39169949 PMCID: PMC11338018 DOI: 10.3389/fnmol.2024.1430080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
Proinflammatory cytokines are implicated in promoting neurodegeneration in multiple sclerosis (MS) by affecting excitatory and inhibitory transmission at central synapses. Conversely, the synaptic effects of anti-inflammatory molecules remain underexplored, despite their potential neuroprotective properties and their presence in the cerebrospinal fluid (CSF) of patients. In a study involving 184 newly diagnosed relapsing-remitting (RR)-MS patients, we investigated whether CSF levels of the anti-inflammatory interleukin (IL)-10 were linked to disease severity and neurodegeneration measures. Additionally, we examined IL-10 impact on synaptic transmission in striatal medium spiny neurons and its role in counteracting inflammatory synaptopathy induced by IL-1β in female C57BL/6 mice with experimental autoimmune encephalomyelitis (EAE). Our findings revealed a significant positive correlation between IL-10 CSF levels and changes in EDSS (Expanded Disability Status Scale) scores one year after MS diagnosis. Moreover, IL-10 levels in the CSF were positively correlated with volumes of specific subcortical brain structures, such as the nucleus caudate. In both MS patients' CSF and EAE mice striatum, IL-10 and IL-1β expressions were upregulated, suggesting possible antagonistic effects of these cytokines. Notably, IL-10 exhibited the ability to decrease glutamate transmission, increase GABA transmission in the striatum, and reverse IL-1β-induced abnormal synaptic transmission in EAE. In conclusion, our data suggest that IL-10 exerts direct neuroprotective effects in MS patients by modulating both excitatory and inhibitory transmission and attenuating IL-1β-induced inflammatory synaptopathy. These findings underscore the potential therapeutic significance of IL-10 in mitigating neurodegeneration in MS.
Collapse
Affiliation(s)
- Luana Gilio
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Faculty of Psychology, Uninettuno Telematic International University, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Roma, Italy
| | | | | | - Krizia Sanna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Giovanni Galifi
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ilaria Simonelli
- Clinical Trial Centre Isola Tiberina-Gemelli Isola, Rome, Italy
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
| | - Fabio Buttari
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ettore Dolcetti
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Antonio Bruno
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | | | | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- IRCCS San Raffaele Scientific Institute, University Vita-Salute San Raffaele, Milan, Italy
| | - Diego Centonze
- Neurology Unit, IRCCS Neuromed, Pozzilli, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Roma, Italy
| |
Collapse
|
13
|
Akatani R, Chihara N, Hara A, Tsuji A, Koto S, Kobayashi K, Toda T, Matsumoto R. Interleukin-6 Signaling Blockade Induces Regulatory Plasmablasts in Neuromyelitis Optica Spectrum Disorder. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200266. [PMID: 38889374 PMCID: PMC11188987 DOI: 10.1212/nxi.0000000000200266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/17/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND OBJECTIVES Interleukin-6 receptor antibodies (IL-6R Abs), including satralizumab, are increasingly used to prevent relapse for neuromyelitis optica spectrum disorder (NMOSD). However, the detailed mechanism of action of this treatment on the lymphocyte phenotype remains unclear. This study focused on B cells in patients with NMOSD, hypothesizing that IL-6R Ab enables B cells to acquire regulatory functions by producing the anti-inflammatory cytokine IL-10. METHODS Peripheral blood mononuclear cells were stimulated in vitro to induce the expansion of B-cell subsets, double-negative B cells (DNs; CD19+ IgD-, CD27-) and plasmablasts (PBs; CD19+, CD27hi, CD38hi). Whole B cells, DNs, or PBs were isolated after culture with IL-6R Ab, and IL-10 expression was quantified using quantitative PCR and a cytometric bead array. RNA sequencing was performed to identify the marker of regulatory PBs induced by IL-6R Ab. RESULTS DNs and PBs were observed to expand in patients with NMSOD during the acute attacks. In the in vitro model, IL-6R Ab increased IL-10 expression in B cells. Notably, IL-10 expression increased in PBs but not in DNs. Using RNA sequencing, CD200 was identified as a marker of regulatory PBs among the differentially expressed upregulated genes. CD200+ PBs produced more IL-10 than CD200- PBs. Furthermore, patients with NMOSD who received satralizumab had a higher proportion of CD200+ PBs than patients during the acute attacks. DISCUSSION Treatment with IL-6 signaling blockade elicited a regulatory phenotype in B cells and PBs. CD200+ PBs may be a marker of treatment responsiveness in the context of NMOSD pathophysiology.
Collapse
Affiliation(s)
- Ritsu Akatani
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Norio Chihara
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Atsushi Hara
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Asato Tsuji
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Shusuke Koto
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Kazuhiro Kobayashi
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Tatsushi Toda
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| | - Riki Matsumoto
- From the Division of Neurology (R.A., N.C., A.H., A.T., S.K., R.M.); Division of Molecular Brain Science (K.K.), Kobe University Graduate School of Medicine; and Department of Neurology (T.T.), Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
14
|
Williams CG, Moreira ML, Asatsuma T, Lee HJ, Li S, Barrera I, Murray E, Soon MSF, Engel JA, Khoury DS, Le S, Wanrooy BJ, Schienstock D, Alexandre YO, Skinner OP, Joseph R, Beattie L, Mueller SN, Chen F, Haque A. Plasmodium infection induces phenotypic, clonal, and spatial diversity among differentiating CD4 + T cells. Cell Rep 2024; 43:114317. [PMID: 38848213 DOI: 10.1016/j.celrep.2024.114317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/21/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
Naive CD4+ T cells must differentiate in order to orchestrate immunity to Plasmodium, yet understanding of their emerging phenotypes, clonality, spatial distributions, and cellular interactions remains incomplete. Here, we observe that splenic polyclonal CD4+ T cells differentiate toward T helper 1 (Th1) and T follicular helper (Tfh)-like states and exhibit rarer phenotypes not elicited among T cell receptor (TCR) transgenic counterparts. TCR clones present at higher frequencies exhibit Th1 skewing, suggesting that variation in major histocompatibility complex class II (MHC-II) interaction influences proliferation and Th1 differentiation. To characterize CD4+ T cell interactions, we map splenic microarchitecture, cellular locations, and molecular interactions using spatial transcriptomics at near single-cell resolution. Tfh-like cells co-locate with stromal cells in B cell follicles, while Th1 cells in red pulp co-locate with activated monocytes expressing multiple chemokines and MHC-II. Spatial mapping of individual transcriptomes suggests that proximity to chemokine-expressing monocytes correlates with stronger effector phenotypes in Th1 cells. Finally, CRISPR-Cas9 gene disruption reveals a role for CCR5 in promoting clonal expansion and Th1 differentiation. A database of cellular locations and interactions is presented: https://haquelab.mdhs.unimelb.edu.au/spatial_gui/.
Collapse
Affiliation(s)
- Cameron G Williams
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Marcela L Moreira
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Takahiro Asatsuma
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Hyun Jae Lee
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Shihan Li
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Irving Barrera
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Evan Murray
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Megan S F Soon
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - Jessica A Engel
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD 4006, Australia
| | - David S Khoury
- Kirby Institute, University of New South Wales, Kensington, NSW 2052, Australia
| | - Shirley Le
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Brooke J Wanrooy
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Dominick Schienstock
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Yannick O Alexandre
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Oliver P Skinner
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Rainon Joseph
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Ashraful Haque
- Department of Microbiology and Immunology, University of Melbourne, located at the Peter Doherty Institute for Infection and Immunity, Parkville, VIC 3000, Australia.
| |
Collapse
|
15
|
Baumgarth N, Prieto AC, Luo Z, Kulaga H. B cells modulate lung antiviral inflammatory responses via the neurotransmitter acetylcholine. RESEARCH SQUARE 2024:rs.3.rs-4421566. [PMID: 38978583 PMCID: PMC11230464 DOI: 10.21203/rs.3.rs-4421566/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The rapid onset of innate immune defenses is critical for early control of viral replication in an infected host, yet it can also lead to irreversible tissue damage, especially in the respiratory tract. Intricate regulatory mechanisms must exist that modulate inflammation, while controlling the infection. Here, B cells expressing choline acetyl transferase (ChAT), an enzyme required for production of the metabolite and neurotransmitter acetylcholine (ACh) are identified as such regulators of the immediate early response to influenza A virus. Lung tissue ChAT + B cells are shown to interact with a7 nicotinic Ach receptor-expressing lung interstitial macrophages in mice within 24h of infection to control their production of TNFa, shifting the balance towards reduced inflammation at the cost of enhanced viral replication. Thus, innate-stimulated B cells are key participants of an immediate-early regulatory cascade that controls lung tissue damage after viral infection.
Collapse
|
16
|
Saito Y, Harada A, Ushijima M, Tanaka K, Higuchi R, Baba A, Murakami D, Nutt SL, Nakagawa T, Ohkawa Y, Baba Y. Plasma cell differentiation is regulated by the expression of histone variant H3.3. Nat Commun 2024; 15:5004. [PMID: 38902223 PMCID: PMC11190180 DOI: 10.1038/s41467-024-49375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/04/2024] [Indexed: 06/22/2024] Open
Abstract
The differentiation of B cells into plasma cells is associated with substantial transcriptional and epigenetic remodeling. H3.3 histone variant marks active chromatin via replication-independent nucleosome assembly. However, its role in plasma cell development remains elusive. Herein, we show that during plasma cell differentiation, H3.3 is downregulated, and the deposition of H3.3 and chromatin accessibility are dynamically changed. Blockade of H3.3 downregulation by enforced H3.3 expression impairs plasma cell differentiation in an H3.3-specific sequence-dependent manner. Mechanistically, enforced H3.3 expression inhibits the upregulation of plasma cell-associated genes such as Irf4, Prdm1, and Xbp1 and maintains the expression of B cell-associated genes, Pax5, Bach2, and Bcl6. Concomitantly, sustained H3.3 expression prevents the structure of chromatin accessibility characteristic for plasma cells. Our findings suggest that appropriate H3.3 expression and deposition control plasma cell differentiation.
Collapse
Affiliation(s)
- Yuichi Saito
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihito Harada
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Miho Ushijima
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryota Higuchi
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akemi Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Daisuke Murakami
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3050, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Takashi Nakagawa
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
17
|
Figueiredo Galvao HB, Lieu M, Moodley S, Diep H, Jelinic M, Bobik A, Sobey CG, Drummond GR, Vinh A. Depletion of follicular B cell-derived antibody secreting cells does not attenuate angiotensin II-induced hypertension or vascular compliance. Front Cardiovasc Med 2024; 11:1419958. [PMID: 38883991 PMCID: PMC11176447 DOI: 10.3389/fcvm.2024.1419958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Marginal zone and follicular B cells are known to contribute to the development of angiotensin II-induced hypertension in mice, but the effector function(s) mediating this effect (e.g., antigen presentation, antibody secretion and/or cytokine production) are unknown. B cell differentiation into antibody secreting cells (ASCs) requires the transcription factor Blimp-1. Here, we studied mice with a Blimp-1 deficiency in follicular B cells to evaluate whether antibody secretion underlies the pro-hypertensive action of B cells. Methods 10- to 14-week-old male follicular B cell Blimp-1 knockout (FoB-Blimp-1-KO) and floxed control mice were subcutaneously infused with angiotensin II (0.7 mg/kg/d) or vehicle (0.1% acetic acid in saline) for 28 days. BP was measured by tail-cuff plethysmography or radiotelemetry. Pulse wave velocity was measured by ultrasound. Aortic collagen was quantified by Masson's trichrome staining. Cell types and serum antibodies were quantified by flow cytometry and a bead-based multiplex assay, respectively. Results In control mice, angiotensin II modestly increased serum IgG3 levels and markedly increased BP, cardiac hypertrophy, aortic stiffening and fibrosis. FoB-Blimp-1-KO mice exhibited impaired IgG1, IgG2a and IgG3 production despite having comparable numbers of B cells and ASCs to control mice. Nevertheless, FoB-Blimp-1-KO mice still developed hypertension, cardiac hypertrophy, aortic stiffening and fibrosis following angiotensin II infusion. Conclusions Inhibition of follicular B cell differentiation into ASCs did not protect against angiotensin II-induced hypertension or vascular compliance. Follicular B cell functions independent of their differentiation into ASCs and ability to produce high-affinity antibodies, or other B cell subtypes, are likely to be involved in angiotensin II-induced hypertension.
Collapse
Affiliation(s)
- Hericka Bruna Figueiredo Galvao
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Maggie Lieu
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Seyuri Moodley
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Henry Diep
- Victorian Heart Institute, Monash University, Clayton, VIC, Australia
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Alexander Bobik
- Baker Heart and Diabetes Institute, Prahran, VIC, Australia
- Department of Immunology, Monash University, Clayton, VIC, Australia
- Center for Inflammatory Diseases, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, VIC, Australia
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, VIC, Australia
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research (CCBDR), La Trobe Institute of Medical Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Elias C, Chen C, Cherukuri A. Regulatory B Cells in Solid Organ Transplantation: From Immune Monitoring to Immunotherapy. Transplantation 2024; 108:1080-1089. [PMID: 37779239 PMCID: PMC10985051 DOI: 10.1097/tp.0000000000004798] [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] [Indexed: 10/03/2023]
Abstract
Regulatory B cells (Breg) modulate the immune response in diverse disease settings including transplantation. Despite the lack of a specific phenotypic marker or transcription factor, their significance in transplantation is underscored by their ability to prolong experimental allograft survival, the possibility for their clinical use as immune monitoring tools, and the exciting prospect for them to form the basis for cell therapy. Interleukin (IL)-10 expression remains the most widely used marker for Breg. Several Breg subsets with distinct phenotypes that express this "signature Breg cytokine" have been described in mice and humans. Although T-cell immunoglobulin and mucin family-1 is the most inclusive and functional marker that accounts for murine Breg with disparate mechanisms of action, the significance of T-cell immunoglobulin and mucin family-1 as a marker for Breg in humans still needs to be explored. Although the primary focus of this review is the role of Breg in clinical transplantation, the net modulatory effect of B cells on the immune response and clinical outcomes is the result of the balancing functions of both Breg and effector B cells. Supporting this notion, B-cell IL-10/tumor necrosis factor α ratio is shown to predict immunologic reactivity and clinical outcomes in kidney and liver transplantation. Assessment of Breg:B effector balance using their IL-10/tumor necrosis factor α ratio may identify patients that require more immunosuppression and provide mechanistic insights into potential therapies. In summary, current advances in our understanding of murine and human Breg will pave way for future definitive clinical studies aiming to test them for immune monitoring and as therapeutic targets.
Collapse
Affiliation(s)
- Charbel Elias
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chuxiao Chen
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Aravind Cherukuri
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Renal and Electrolyte Division, Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Li Q, Yang C, Liu C, Zhang Y, An N, Ma X, Zheng Y, Cui X, Li Q. The circulating IL-35 + regulatory B cells are associated with thyroid associated opthalmopathy. Immun Inflamm Dis 2024; 12:e1304. [PMID: 38804861 PMCID: PMC11131934 DOI: 10.1002/iid3.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/26/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Thyroid-associated ophthalmopathy (TAO) is the most common orbital disease in adults, potentially leading to disfigurement and visual impairment. However, the causes of TAO are not fully understood. IL-35+B cells are a newly identified regulatory B cells (Bregs) in maintaining immune balance in various autoimmune diseases. Yet, the influence of IL-35+Bregs in TAO remains unexplored. METHODS This study enrolled 36 healthy individuals and 14 TAO patients. We isolated peripheral blood mononuclear cells and stimulated them with IL-35 and CpG for 48 h. Flow cytometry was used to measure the percentages of IL-35+Bregs. RESULTS The percentage of circulating IL-35+Bregs was higher in TAO patients, and this increase correlated positively with disease activity. IL-35 significantly increased the generation of IL-35+Bregs in healthy individuals. However, B cells from TAO patients exhibited potential impairment in transitioning into IL-35+Breg phenotype under IL-35 stimulation. CONCLUSIONS Our results suggest a potential role of IL-35+Bregs in the development of TAO, opening new avenues for understanding disease mechanisms and developing therapeutic approaches.
Collapse
Affiliation(s)
- Qian Li
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Cuixia Yang
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Cheng Liu
- Medical Science Research Institution of Ningxia Hui Autonomous RegionMedical Sci‐Tech Research Center of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Yuehui Zhang
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Ningyu An
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Xiumei Ma
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Yang Zheng
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Xiaomin Cui
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| | - Qian Li
- Department of Ophthalmology, People's Hospital of Ningxia Hui Autonomous RegionThe Third Affiliated Clinical College of Ningxia Medical UniversityYinchuanNingxia Hui Autonomous RegionChina
| |
Collapse
|
20
|
Hu F, Shi L, Liu X, Chen Y, Zhang X, Jia Y, Liu X, Guo J, Zhu H, Liu H, Xu L, Li Y, Wang P, Fang X, Xue J, Xie Y, Wei C, Song J, Zheng X, Liu YY, Li Y, Ren L, Xu D, Lu L, Qiu X, Mu R, He J, Wang M, Zhang X, Liu W, Li Z. Proinflammatory phenotype of B10 and B10pro cells elicited by TNF-α in rheumatoid arthritis. Ann Rheum Dis 2024; 83:576-588. [PMID: 38302261 DOI: 10.1136/ard-2023-224878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVES B10 and B10pro cells suppress immune responses via secreting interleukin (IL)-10. However, their regulators and underlying mechanisms, especially in human autoimmune diseases, are elusive. This study aimed to address these questions in rheumatoid arthritis (RA), one of the most common highly disabling autoimmune diseases. METHODS The frequencies and functions of B10 and B10pro cells in healthy individuals and patients with RA were first analysed. The effects of proinflammatory cytokines, particularly tumour necrosis factor (TNF)-α on the quantity, stability and pathogenic phenotype of these cells, were then assessed in patients with RA before and after anti-TNF therapy. The underlying mechanisms were further investigated by scRNA-seq database reanalysis, transcriptome sequencing, TNF-α-/- and B cell-specific SHIP-1-/- mouse disease model studies. RESULTS TNF-α was a key determinant for B10 cells. TNF-α elicited the proinflammatory feature of B10 and B10pro cells by downregulating IL-10, and upregulating interferon-γ and IL-17A. In patients with RA, B10 and B10pro cells were impaired with exacerbated proinflammatory phenotype, while anti-TNF therapy potently restored their frequencies and immunosuppressive functions, consistent with the increased B10 cells in TNF-α-/- mice. Mechanistically, TNF-α diminished B10 and B10pro cells by inhibiting their glycolysis and proliferation. TNF-α also regulated the phosphatidylinositol phosphate signalling of B10 and B10pro cells and dampened the expression of SHIP-1, a dominant phosphatidylinositol phosphatase regulator of these cells. CONCLUSIONS TNF-α provoked the proinflammatory phenotype of B10 and B10pro cells by disturbing SHIP-1 in RA, contributing to the disease development. Reinstating the immunosuppressive property of B10 and B10pro cells might represent novel therapeutic approaches for RA.
Collapse
Affiliation(s)
- 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
| | - Lianjie Shi
- Department of Rheumatology and Immunology, Peking University Shougang Hospital, Beijing, China
| | - Xiaohang Liu
- State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Yingjia Chen
- State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, China
| | - Xia Zhang
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuan Jia
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), 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
| | - Jianping Guo
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Huaqun Zhu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Hongjiang Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Liling Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yingni Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Ping Wang
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xiangyu Fang
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jimeng Xue
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yang Xie
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Chaonan Wei
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Jing Song
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Xi Zheng
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Yan-Ying Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuhui Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Limin Ren
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liwei Lu
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Rong Mu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, China
| | - Jing He
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, China Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Institute for Immunology, Tsinghua University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Zhanguo Li
- 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
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
21
|
Tompa A, Faresjö M. Shift in the B cell subsets between children with type 1 diabetes and/or celiac disease. Clin Exp Immunol 2024; 216:36-44. [PMID: 38134245 PMCID: PMC10929695 DOI: 10.1093/cei/uxad136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/17/2023] [Indexed: 12/24/2023] Open
Abstract
Our purpose was to characterize the pattern of B cell subsets in children with a combined diagnosis of type 1 diabetes (T1D) and celiac disease (C) since children with single or double diagnosis of these autoimmune diseases may differ in peripheral B cell subset phenotype patterns. B cells were analyzed with flow cytometry for the expression of differentiation/maturation markers to identify transitional, naive, and memory B cells. Transitional (CD24hiCD38hiCD19+) and memory Bregs (mBregs; CD24hiCD27+CD19+, CD1d+CD27+CD19+, and CD5+CD1d+CD19+) were classified as B cells with regulatory capacity. Children with a combined diagnosis of T1D and C showed a pattern of diminished peripheral B cell subsets. The B cells compartment in children with combined diagnosis had higher percentages of memory B subsets and Bregs, including activated subsets, compared to children with either T1D or C. Children with combined diagnosis had a lower percentage of naive B cells (CD27-CD19+; IgD+CD19+) and an increased percentage of memory B cells (CD27+CD19+; IgD-CD19+). A similar alteration was seen among the CD39+ expressing naive and memory B cells. Memory Bregs (CD1d+CD27+CD19+) were more frequent, contrary to the lower percentage of CD5+ transitional Bregs in children with a combined diagnosis. In children with either T1D or C, the peripheral B cell compartment was dominated by naive cells. Differences in the pattern of heterogeneous peripheral B cell repertoire subsets reflect a shifting in the B cell compartment between children with T1D and/or C. This is an immunological challenge of impact on the pathophysiology of these autoimmune diseases.
Collapse
Affiliation(s)
- Andrea Tompa
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Division of Diagnostics, Region Jönköping County, Jönköping, Sweden
| | - Maria Faresjö
- Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden
- Department of Life Sciences, Division of Systems and Synthetic Biology, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
22
|
Stock AJ, Gonzalez Paredes P, de Almeida LP, Kosanke SD, Chetlur S, Budde H, Wakenight P, Zwingman TA, Rosen AB, Allenspach EJ, Millen KJ, Buckner JH, Rawlings DJ, Gorman JA. The IFIH1-A946T risk variant promotes diabetes in a sex-dependent manner. Front Immunol 2024; 15:1349601. [PMID: 38487540 PMCID: PMC10937421 DOI: 10.3389/fimmu.2024.1349601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/14/2024] [Indexed: 03/17/2024] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic islet β-cells are attacked by the immune system, resulting in insulin deficiency and hyperglycemia. One of the top non-synonymous single-nucleotide polymorphisms (SNP) associated with T1D is in the interferon-induced helicase C domain-containing protein 1 (IFIH1), which encodes an anti-viral cytosolic RNA sensor. This SNP results in an alanine to threonine substitution at amino acid 946 (IFIH1A946T) and confers an increased risk for several autoimmune diseases, including T1D. We hypothesized that the IFIH1A946T risk variant, (IFIH1R) would promote T1D pathogenesis by stimulating type I interferon (IFN I) signaling leading to immune cell alterations. To test this, we developed Ifih1R knock-in mice on the non-obese diabetic (NOD) mouse background, a spontaneous T1D model. Our results revealed a modest increase in diabetes incidence and insulitis in Ifih1R compared to non-risk Ifih1 (Ifih1NR) mice and a significant acceleration of diabetes onset in Ifih1R females. Ifih1R mice exhibited a significantly enhanced interferon stimulated gene (ISG) signature compared to Ifih1NR, indicative of increased IFN I signaling. Ifih1R mice exhibited an increased frequency of plasma cells as well as tissue-dependent changes in the frequency and activation of CD8+ T cells. Our results indicate that IFIH1R may contribute to T1D pathogenesis by altering the frequency and activation of immune cells. These findings advance our knowledge on the connection between the rs1990760 variant and T1D. Further, these data are the first to demonstrate effects of Ifih1R in NOD mice, which will be important to consider for the development of therapeutics for T1D.
Collapse
Affiliation(s)
- Amanda J. Stock
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology, Oklahoma City, OK, United States
| | - Pierina Gonzalez Paredes
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology, Oklahoma City, OK, United States
| | | | - Stanley D. Kosanke
- Heartland Veterinary Pathology Services, PLLC, Edmond, OK, United States
| | - Srinivaas Chetlur
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology, Oklahoma City, OK, United States
| | - Hannah Budde
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology, Oklahoma City, OK, United States
| | - Paul Wakenight
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | - Theresa A. Zwingman
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
| | - Aaron B.I. Rosen
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapies, Seattle, WA, United States
| | - Eric J. Allenspach
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapies, Seattle, WA, United States
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States
| | - Kathleen J. Millen
- Seattle Children’s Research Institute, Center for Integrative Brain Research, Seattle, WA, United States
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States
| | - Jane H. Buckner
- Benaroya Research Institute at Virginia Mason, Center for Translational Immunology, Seattle, WA, United States
| | - David J. Rawlings
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapies, Seattle, WA, United States
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, United States
- Department of Immunology, School of Medicine, University of Washington, Seattle, WA, United States
| | - Jacquelyn A. Gorman
- Oklahoma Medical Research Foundation, Arthritis & Clinical Immunology, Oklahoma City, OK, United States
| |
Collapse
|
23
|
Jardou M, Brossier C, Marquet P, Picard N, Druilhe A, Lawson R. Solid organ transplantation and gut microbiota: a review of the potential immunomodulatory properties of short-chain fatty acids in graft maintenance. Front Cell Infect Microbiol 2024; 14:1342354. [PMID: 38476165 PMCID: PMC10927761 DOI: 10.3389/fcimb.2024.1342354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Transplantation is the treatment of choice for several end-stage organ defects: it considerably improves patient survival and quality of life. However, post-transplant recipients may experience episodes of rejection that can favor or ultimately lead to graft loss. Graft maintenance requires a complex and life-long immunosuppressive treatment. Different immunosuppressive drugs (i.e., calcineurin inhibitors, glucocorticoids, biological immunosuppressive agents, mammalian target of rapamycin inhibitors, and antiproliferative or antimetabolic agents) are used in combination to mitigate the immune response against the allograft. Unfortunately, the use of these antirejection agents may lead to opportunistic infections, metabolic (e.g., post-transplant diabetes mellitus) or cardiovascular (e.g., arterial hypertension) disorders, cancer (e.g., non-Hodgkin lymphoma) and other adverse effects. Lately, immunosuppressive drugs have also been associated with gut microbiome alterations, known as dysbiosis, and were shown to affect gut microbiota-derived short-chain fatty acids (SCFA) production. SCFA play a key immunomodulatory role in physiological conditions, and their impairment in transplant patients could partly counterbalance the effect of immunosuppressive drugs leading to the activation of deleterious pathways and graft rejection. In this review, we will first present an overview of the mechanisms of graft rejection that are prevented by the immunosuppressive protocol. Next, we will explain the dynamic changes of the gut microbiota during transplantation, focusing on SCFA. Finally, we will describe the known functions of SCFA in regulating immune-inflammatory reactions and discuss the impact of SCFA impairment in immunosuppressive drug treated patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Roland Lawson
- National Institute of Health and Medical Research (FRANCE) (INSERM), Univ. Limoges, Pharmacology & Transplantation, U1248, Limoges, France
| |
Collapse
|
24
|
Veh J, Ludwig C, Schrezenmeier H, Jahrsdörfer B. Regulatory B Cells-Immunopathological and Prognostic Potential in Humans. Cells 2024; 13:357. [PMID: 38391970 PMCID: PMC10886933 DOI: 10.3390/cells13040357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
The aim of the following review is to shed light on the putative role of regulatory B cells (Bregs) in various human diseases and highlight their potential prognostic and therapeutic relevance in humans. Regulatory B cells are a heterogeneous group of B lymphocytes capable of suppressing inflammatory immune reactions. In this way, Bregs contribute to the maintenance of tolerance and immune homeostasis by limiting ongoing immune reactions temporally and spatially. Bregs play an important role in attenuating pathological inflammatory reactions that can be associated with transplant rejection, graft-versus-host disease, autoimmune diseases and allergies but also with infectious, neoplastic and metabolic diseases. Early studies of Bregs identified IL-10 as an important functional molecule, so the IL-10-secreting murine B10 cell is still considered a prototype Breg, and IL-10 has long been central to the search for human Breg equivalents. However, over the past two decades, other molecules that may contribute to the immunosuppressive function of Bregs have been discovered, some of which are only present in human Bregs. This expanded arsenal includes several anti-inflammatory cytokines, such as IL-35 and TGF-β, but also enzymes such as CD39/CD73, granzyme B and IDO as well as cell surface proteins including PD-L1, CD1d and CD25. In summary, the present review illustrates in a concise and comprehensive manner that although human Bregs share common functional immunosuppressive features leading to a prominent role in various human immunpathologies, they are composed of a pool of different B cell types with rather heterogeneous phenotypic and transcriptional properties.
Collapse
Affiliation(s)
- Johanna Veh
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, 89081 Ulm, Germany
| | - Carolin Ludwig
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, 89081 Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute for Transfusion Medicine, Ulm University Hospitals and Clinics, 89081 Ulm, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Donation Service Baden-Württemberg-Hessen, 89081 Ulm, Germany
| |
Collapse
|
25
|
Sanli E, Sirin NG, Kucukali CI, Baykan B, Ulusoy CA, Bebek N, Yilmaz V, Tuzun E. Peripheral blood regulatory B and T cells are decreased in patients with focal epilepsy. J Neuroimmunol 2024; 387:578287. [PMID: 38241950 DOI: 10.1016/j.jneuroim.2024.578287] [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: 10/28/2023] [Revised: 12/12/2023] [Accepted: 01/06/2024] [Indexed: 01/21/2024]
Abstract
Patients with focal epilepsy of unknown cause (FEoUC) may display T cell infiltration in post-surgery brain specimens and increased serum levels of pro-inflammatory cytokines produced by B and T cells, indicating potential involvement of adaptive immunity. Our study aimed to investigate the peripheral blood distribution of B and T cell subgroups to find clues supporting the distinct organization of adaptive immunity in FEoUC. Twenty-two patients with FEoUC and 25 age and sex matched healthy individuals were included. Peripheral blood mononuclear cells were immunophenotyped by flow cytometry. Expression levels of anti-inflammatory cytokines and FOXP3 were measured by real-time PCR. Carboxyfluorescein succinimidyl ester (CFSE) proliferation assay was conducted using CD4+ T cells. Patients with FEoUC showed significantly decreased regulatory B (Breg), B1a, plasmablast and regulatory T (Treg) cell percentages, and increased switched memory B and Th17 cell ratios. Moreover, CD4+CD25+CD49d- Tregs of FEoUC patients displayed significantly reduced TGFB1 and FOXP3, but increased IL10 gene expression levels. CD4+ helper T cells of patients with FEoUC gave more exaggerated proliferation responses to phytohemagglutinin, anti-CD3 and anti-CD28 stimulation. Patients with FEoUC display increased effector lymphocyte, decreased regulatory lymphocyte ratios, and impaired Treg function and enhanced lymphocyte proliferation capacity. Overall, this pro-inflammatory phenotype lends support to the involvement of adaptive immunity in FEoUC.
Collapse
Affiliation(s)
- Elif Sanli
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey; Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey
| | - Nermin Gorkem Sirin
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cem Ismail Kucukali
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Betul Baykan
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey; Department of Neurology, EMAR Medical Center, Istanbul, Turkey
| | - Canan Aysel Ulusoy
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Nerses Bebek
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Vuslat Yilmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| | - Erdem Tuzun
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
26
|
Yu Y, Lu C, Yu W, Lei Y, Sun S, Liu P, Bai F, Chen Y, Chen J. B Cells Dynamic in Aging and the Implications of Nutritional Regulation. Nutrients 2024; 16:487. [PMID: 38398810 PMCID: PMC10893126 DOI: 10.3390/nu16040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Aging negatively affects B cell production, resulting in a decrease in B-1 and B-2 cells and impaired antibody responses. Age-related B cell subsets contribute to inflammation. Investigating age-related alterations in the B-cell pool and developing targeted therapies are crucial for combating autoimmune diseases in the elderly. Additionally, optimal nutrition, including carbohydrates, amino acids, vitamins, and especially lipids, play a vital role in supporting immune function and mitigating the age-related decline in B cell activity. Research on the influence of lipids on B cells shows promise for improving autoimmune diseases. Understanding the aging B-cell pool and considering nutritional interventions can inform strategies for promoting healthy aging and reducing the age-related disease burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100091, China; (Y.Y.)
| |
Collapse
|
27
|
Goldmann O, Nwofor OV, Chen Q, Medina E. Mechanisms underlying immunosuppression by regulatory cells. Front Immunol 2024; 15:1328193. [PMID: 38380317 PMCID: PMC10876998 DOI: 10.3389/fimmu.2024.1328193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/16/2024] [Indexed: 02/22/2024] Open
Abstract
Regulatory cells, such as regulatory T cells (Tregs), regulatory B cells (Bregs), and myeloid-derived suppressor cells (MDSCs), play a crucial role in preserving immune tolerance and controlling immune responses during infections to prevent excessive immune activation. However, pathogens have developed strategies to hijack these regulatory cells to decrease the overall effectiveness of the immune response and persist within the host. Consequently, therapeutic targeting of these immunosuppressive mechanisms during infection can reinvigorate the immune response and improve the infection outcome. The suppressive mechanisms of regulatory cells are not only numerous but also redundant, reflecting the complexity of the regulatory network in modulating the immune responses. The context of the immune response, such as the type of pathogen or tissue involved, further influences the regulatory mechanisms involved. Examples of these immunosuppressive mechanisms include the production of inhibitory cytokines such as interleukin 10 (IL-10) and transforming growth factor beta (TGF-β) that inhibit the production of pro-inflammatory cytokines and dampen the activation and proliferation of effector T cells. In addition, regulatory cells utilize inhibitory receptors like cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) to engage with their respective effector cells, thereby suppressing their function. An alternative approach involves the modulation of metabolic reprogramming in effector immune cells to limit their activation and proliferation. In this review, we provide an overview of the major mechanisms mediating the immunosuppressive effect of the different regulatory cell subsets in the context of infection.
Collapse
Affiliation(s)
| | | | | | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
28
|
Stock AJ, Gonzalez-Paredes P, Previato de Almeida L, Kosanke SD, Chetlur S, Budde H, Wakenight P, Zwingman TA, Rosen AB, Allenspach E, Millen KJ, Buckner JH, Rawlings DJ, Gorman JA. The IFIH1-A946T risk variant promotes diabetes in a sex-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576482. [PMID: 38328221 PMCID: PMC10849491 DOI: 10.1101/2024.01.20.576482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic islet β-cells are attacked by the immune system, resulting in insulin deficiency and hyperglycemia. One of the top non-synonymous single-nucleotide polymorphisms (SNP) associated with T1D is in the interferon-induced helicase C domain-containing protein 1 ( IFIH1 ), which encodes an anti-viral cytosolic RNA sensor. This SNP results in an alanine to threonine substitution at amino acid 946 (IFIH1 A946T ) and confers an increased risk for several autoimmune diseases, including T1D. We hypothesized that the IFIH1 A946T risk variant, ( IFIH1 R ) would promote T1D pathogenesis by stimulating type I interferon (IFN I) signaling leading to immune cell alterations. To test this, we developed Ifih1 R knock-in mice on the non-obese diabetic (NOD) mouse background, a spontaneous T1D model. Our results revealed a modest increase in diabetes incidence and insulitis in Ifih1 R compared to non-risk Ifih1 ( Ifih1 NR ) mice and a significant acceleration of diabetes onset in Ifih1 R females. Ifih1 R mice exhibited a significantly enhanced interferon stimulated gene (ISG) signature compared to Ifih1 NR , indicative of increased IFN I signaling. Ifih1 R mice exhibited an increased frequency of plasma cells as well as tissue-dependent changes in the frequency and activation of CD8 + T cells. Our results indicate that IFIH1 R may contribute to T1D pathogenesis by altering the frequency and activation of immune cells. These findings advance our knowledge on the connection between the rs1990760 variant and T1D. Further, these data are the first to demonstrate effects of Ifih1 R in NOD mice, which will be important to consider for the development of therapeutics for T1D.
Collapse
|
29
|
Kliem CV, Schaub B. The role of regulatory B cells in immune regulation and childhood allergic asthma. Mol Cell Pediatr 2024; 11:1. [PMID: 38172451 PMCID: PMC10764675 DOI: 10.1186/s40348-023-00174-2] [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: 09/16/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND As the most common chronic disease in childhood, asthma displays a major public health problem worldwide with the incidence of those affected rising. As there is currently no cure for allergic asthma, it is mandatory to get a better understanding of the underlying molecular mechanism. MAIN BODY By producing IgE antibodies upon allergen contact, B cells play a pivotal role in allergic asthma. Besides that, IL-10-secreting B cell subsets, namely regulatory B cells (Bregs), are reported in mice and humans to play a role in allergic asthma. In humans, several Breg subsets with distinct phenotypic and functional properties are identified among B cells at different maturational and differentiation stages that exert anti-inflammatory functions by expressing several suppressor molecules. Emerging research has focused on the role of Bregs in allergic asthma as well as their role for future diagnostic and preventive strategies. CONCLUSION Knowledge about the exact function of human Bregs in allergic asthma is still very limited. This review aims to summarize the current knowledge on Bregs. We discuss different human Breg subsets, several ways of Breg induction as well as the mechanisms through which they exert immunoregulatory functions, and their role in (childhood) allergic asthma.
Collapse
Affiliation(s)
- Caroline Vanessa Kliem
- Pediatric Allergology, Department of Pediatrics, Dr. Von Hauner Children´S Hospital, University Hospital, Lindwurmstraße 4, 80337, LMU, Munich, Germany
| | - Bianca Schaub
- Pediatric Allergology, Department of Pediatrics, Dr. Von Hauner Children´S Hospital, University Hospital, Lindwurmstraße 4, 80337, LMU, Munich, Germany.
- Member of German Center for Lung Research - DZL, LMU, Munich, Germany.
- Member of German Center for Child and Adolescent Health-DZKJ, LMU, Munich, Germany.
| |
Collapse
|
30
|
Zheremyan EA, Ustiugova AS, Karamushka NM, Uvarova AN, Stasevich EM, Bogolyubova AV, Kuprash DV, Korneev KV. Breg-Mediated Immunoregulation in the Skin. Int J Mol Sci 2024; 25:583. [PMID: 38203754 PMCID: PMC10778726 DOI: 10.3390/ijms25010583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Wound healing is a complex process involving a coordinated series of events aimed at restoring tissue integrity and function. Regulatory B cells (Bregs) are a subset of B lymphocytes that play an essential role in fine-tuning immune responses and maintaining immune homeostasis. Recent studies have suggested that Bregs are important players in cutaneous immunity. This review summarizes the current understanding of the role of Bregs in skin immunity in health and pathology, such as diabetes, psoriasis, systemic sclerosis, cutaneous lupus erythematosus, cutaneous hypersensitivity, pemphigus, and dermatomyositis. We discuss the mechanisms by which Bregs maintain tissue homeostasis in the wound microenvironment through the promotion of angiogenesis, suppression of effector cells, and induction of regulatory immune cells. We also mention the potential clinical applications of Bregs in promoting wound healing, such as the use of adoptive Breg transfer.
Collapse
Affiliation(s)
- Elina A. Zheremyan
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alina S. Ustiugova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Nina M. Karamushka
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Aksinya N. Uvarova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ekaterina M. Stasevich
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | - Dmitry V. Kuprash
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Kirill V. Korneev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- National Research Center for Hematology, 125167 Moscow, Russia
| |
Collapse
|
31
|
Daamen AR, Alajoleen RM, Grammer AC, Luo XM, Lipsky PE. Single-cell RNA sequencing analysis reveals the heterogeneity of IL-10 producing regulatory B cells in lupus-prone mice. Front Immunol 2023; 14:1282770. [PMID: 38155972 PMCID: PMC10752970 DOI: 10.3389/fimmu.2023.1282770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023] Open
Abstract
Introduction B cells can have both pathogenic and protective roles in autoimmune diseases, including systemic lupus erythematosus (SLE). Deficiencies in the number or immunosuppressive function of IL-10 producing regulatory B cells (Bregs) can cause exacerbated autoimmune inflammation. However, the exact role of Bregs in lupus pathogenesis has not been elucidated. Methods We carried out gene expression analysis by scRNA-seq to characterize differences in splenic Breg subsets and molecular profiles through stages of disease progression in lupus-prone mice. Transcriptome-based changes in Bregs from mice with active disease were confirmed by phenotypic analysis. Results We found that a loss of marginal zone (MZ) lineage Bregs, an increase in plasmablast/plasma cell (PB-PC) lineage Bregs, and overall increases in inflammatory gene signatures were characteristic of active disease as compared to Bregs from the pre-disease stage. However, the frequencies of both MZ Bregs and PB-PCs expressing IL-10 were significantly decreased in active-disease mice. Conclusion Overall, we have identified changes to the repertoire and transcriptional landscape of Breg subsets associated with active disease that provide insights into the role of Bregs in lupus pathogenesis. These results could inform the design of Breg-targeted therapies and interventions to restore Breg suppressive function in autoimmunity.
Collapse
Affiliation(s)
- Andrea R. Daamen
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - Razan M. Alajoleen
- Department of Biomedical Sciences and Pathology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Amrie C. Grammer
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - Xin M. Luo
- Department of Biomedical Sciences and Pathology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Peter E. Lipsky
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| |
Collapse
|
32
|
Neziraj T, Siewert L, Pössnecker E, Pröbstel AK. Therapeutic targeting of gut-originating regulatory B cells in neuroinflammatory diseases. Eur J Immunol 2023; 53:e2250033. [PMID: 37624875 DOI: 10.1002/eji.202250033] [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: 02/06/2023] [Revised: 05/29/2023] [Accepted: 08/23/2023] [Indexed: 08/27/2023]
Abstract
Regulatory B cells (Bregs) are immunosuppressive cells that support immunological tolerance by the production of IL-10, IL-35, and TGF-β. Bregs arise from different developmental stages in response to inflammatory stimuli. In that regard, mounting evidence points towards a direct influence of gut microbiota on mucosal B cell development, activation, and regulation in health and disease. While an increasing number of diseases are associated with alterations in gut microbiome (dysbiosis), little is known about the role of microbiota on Breg development and induction in neuroinflammatory disorders. Notably, gut-originating, IL-10- and IgA-producing regulatory plasma cells have recently been demonstrated to egress from the gut to suppress inflammation in the CNS raising fundamental questions about the triggers and functions of mucosal-originating Bregs in systemic inflammation. Advancing our understanding of Bregs in neuroinflammatory diseases could lead to novel therapeutic approaches. Here, we summarize the main aspects of Breg differentiation and functions and evidence about their involvement in neuroinflammatory diseases. Further, we highlight current data of gut-originating Bregs and their microbial interactions and discuss future microbiota-regulatory B cell-targeted therapies in immune-mediated diseases.
Collapse
Affiliation(s)
- Tradite Neziraj
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Lena Siewert
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Elisabeth Pössnecker
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Anne-Katrin Pröbstel
- Department of Neurology, University Hospital of Basel and University of Basel, Basel, Switzerland
- Departments of Biomedicine and Clinical Research, University Hospital of Basel and University of Basel, Basel, Switzerland
- Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital of Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
33
|
Zogorean R, Wirtz S. The yin and yang of B cells in a constant state of battle: intestinal inflammation and inflammatory bowel disease. Front Immunol 2023; 14:1260266. [PMID: 37849749 PMCID: PMC10577428 DOI: 10.3389/fimmu.2023.1260266] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract, defined by a clinical relapse-remitting course. Affecting people worldwide, the origin of IBD is still undefined, arising as a consequence of the interaction between genes, environment, and microbiota. Although the root cause is difficult to identify, data clearly indicate that dysbiosis and pathogenic microbial taxa are connected with the establishment and clinical course of IBD. The composition of the microbiota is shaped by plasma cell IgA secretion and binding, while cytokines such as IL10 or IFN-γ are important fine-tuners of the immune response in the gastrointestinal environment. B cells may also influence the course of inflammation by promoting either an anti-inflammatory or a pro-inflammatory milieu. Here, we discuss IgA-producing B regulatory cells as an anti-inflammatory factor in intestinal inflammation. Moreover, we specify the context of IgA and IgG as players that can potentially participate in mucosal inflammation. Finally, we discuss the role of B cells in mouse infection models where IL10, IgA, or IgG contribute to the outcome of the infection.
Collapse
Affiliation(s)
- Roxana Zogorean
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Bavaria, Germany
| |
Collapse
|
34
|
Zheng H, Cao P, Su Z, Xia L. Insights into the roles of IL-10-producing regulatory B cells in cardiovascular disorders: recent advances and future perspectives. J Leukoc Biol 2023; 114:315-324. [PMID: 37284816 DOI: 10.1093/jleuko/qiad066] [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: 05/22/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 06/08/2023] Open
Abstract
Interleukin-10-producing regulatory B (B10) cells mediate the immunomodulatory functions of biosystems by secreting anti-inflammatory factors, thus playing vital roles in cardiovascular diseases such as viral myocarditis, myocardial infarction, and ischemia-reperfusion injury. However, several challenges hinder B10 cells from regulating the immunoreactivity of organisms in specific cardiovascular diseases, such as atherosclerotic disease. Regarding the regulatory mechanisms of B10 cells, the interplay between B10 cells and the cardiovascular and immune systems is complex and requires clarification. In this study, we summarize the roles of B10 cells in bacterial and aseptic heart injuries, address their regulatory functions in different stages of cardiovascular disorders, and discuss their challenges and opportunities in addressing cardiovascular diseases from bench to bedside.
Collapse
Affiliation(s)
- Huiqin Zheng
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang 212001, China
- International Genome Center, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, China
| | - Pei Cao
- International Genome Center, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, China
- Institute of Medical Immunology, Jiangsu University, No. 438 Jiefang Road, Zhenjiang 212001, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Zhenjiang 212001, China
- Institute of Hematological Disease, Jiangsu University, No. 438 Jiefang Road, Zhenjiang 212001, China
| |
Collapse
|
35
|
Guinn MT, Szuter ES, Yokose T, Ge J, Rosales IA, Chetal K, Sadreyev RI, Cuenca AG, Kreisel D, Sage PT, Russell PS, Madsen JC, Colvin RB, Alessandrini A. Intragraft B cell differentiation during the development of tolerance to kidney allografts is associated with a regulatory B cell signature revealed by single cell transcriptomics. Am J Transplant 2023; 23:1319-1330. [PMID: 37295719 PMCID: PMC11232115 DOI: 10.1016/j.ajt.2023.05.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/12/2023]
Abstract
Mouse kidney allografts are spontaneously accepted in select, fully mismatched donor-recipient strain combinations, like DBA/2J to C57BL/6 (B6), by natural tolerance. We previously showed accepted renal grafts form aggregates containing various immune cells within 2 weeks posttransplant, referred to as regulatory T cell-rich organized lymphoid structures, which are a novel regulatory tertiary lymphoid organ. To characterize the cells within T cell-rich organized lymphoid structures, we performed single-cell RNA sequencing on CD45+ sorted cells from accepted and rejected renal grafts from 1-week to 6-months posttransplant. Analysis of single-cell RNA sequencing data revealed a shifting from a T cell-dominant to a B cell-rich population by 6 months with an increased regulatory B cell signature. Furthermore, B cells were a greater proportion of the early infiltrating cells in accepted vs rejecting grafts. Flow cytometry of B cells at 20 weeks posttransplant revealed T cell, immunoglobulin domain and mucin domain-1+ B cells, potentially implicating a regulatory role in the maintenance of allograft tolerance. Lastly, B cell trajectory analysis revealed intragraft differentiation from precursor B cells to memory B cells in accepted allografts. In summary, we show a shifting T cell- to B cell-rich environment and a differential cellular pattern among accepted vs rejecting kidney allografts, possibly implicating B cells in the maintenance of kidney allograft acceptance.
Collapse
Affiliation(s)
- Michael Tyler Guinn
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Edward S Szuter
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Takahiro Yokose
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jifu Ge
- Boston's Children Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy A Rosales
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kashish Chetal
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ruslan I Sadreyev
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alex G Cuenca
- Boston's Children Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Kreisel
- Departments of Surgery, Pathology, and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peter T Sage
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul S Russell
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joren C Madsen
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Division of Cardiac Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert B Colvin
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alessandro Alessandrini
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
36
|
Satitsuksanoa P, Iwasaki S, Boersma J, Bel Imam M, Schneider SR, Chang I, van de Veen W, Akdis M. B cells: The many facets of B cells in allergic diseases. J Allergy Clin Immunol 2023; 152:567-581. [PMID: 37247640 DOI: 10.1016/j.jaci.2023.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/30/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
B cells play a key role in our immune system through their ability to produce antibodies, suppress a proinflammatory state, and contribute to central immune tolerance. We aim to provide an in-depth knowledge of the molecular biology of B cells, including their origin, developmental process, types and subsets, and functions. In allergic diseases, B cells are well known to induce and maintain immune tolerance through the production of suppressor cytokines such as IL-10. Similarly, B cells protect against viral infections such as severe acute respiratory syndrome coronavirus 2 that caused the recent coronavirus disease 2019 pandemic. Considering the unique and multifaceted functions of B cells, we hereby provide a comprehensive overview of the current knowledge of B-cell biology and its clinical applications in allergic diseases, organ transplantation, and cancer.
Collapse
Affiliation(s)
- Pattraporn Satitsuksanoa
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland.
| | - Sayuri Iwasaki
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Wageningen University & Research, Wageningen, The Netherlands
| | - Jolien Boersma
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Wageningen University & Research, Wageningen, The Netherlands
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Iris Chang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland; Sean N. Parker Centre for Allergy and Asthma Research, Department of Medicine, Stanford University, Palo Alto, Calif
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Davos, Switzerland.
| |
Collapse
|
37
|
Bugbee E, Wang AA, Gommerman JL. Under the influence: environmental factors as modulators of neuroinflammation through the IL-10/IL-10R axis. Front Immunol 2023; 14:1188750. [PMID: 37600781 PMCID: PMC10435745 DOI: 10.3389/fimmu.2023.1188750] [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: 03/17/2023] [Accepted: 07/12/2023] [Indexed: 08/22/2023] Open
Abstract
The IL-10/IL-10 receptor (IL-10R) axis plays an important role in attenuating neuroinflammation in animal models of Multiple Sclerosis (MS) and increased IL-10 has been associated with a positive response to MS disease modifying therapy. Because environmental factors play an important role in MS susceptibility and disease course, identification of environmental factors that impact the IL-10/IL-10R axis has therapeutic potential. In this review, we provide historical and updated perspectives of how IL-10R signaling impacts neuroinflammation, discuss environmental factors and intestinal microbes with known impacts on the IL-10/IL-10R axis, and provide a hypothetical model for how B cells, via their production of IL-10, may be important in conveying environmental "information" to the inflamed central nervous system.
Collapse
|
38
|
Steinmetz TD, Verstappen GM, Suurmond J, Kroese FGM. Targeting plasma cells in systemic autoimmune rheumatic diseases - Promises and pitfalls. Immunol Lett 2023; 260:44-57. [PMID: 37315847 DOI: 10.1016/j.imlet.2023.06.005] [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: 02/03/2023] [Revised: 05/12/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Plasma cells are the antibody secretors of the immune system. Continuous antibody secretion over years can provide long-term immune protection but could also be held responsible for long-lasting autoimmunity in case of self-reactive plasma cells. Systemic autoimmune rheumatic diseases (ARD) affect multiple organ systems and are associated with a plethora of different autoantibodies. Two prototypic systemic ARDs are systemic lupus erythematosus (SLE) and Sjögren's disease (SjD). Both diseases are characterized by B-cell hyperactivity and the production of autoantibodies against nuclear antigens. Analogues to other immune cells, different subsets of plasma cells have been described. Plasma cell subsets are often defined dependent on their current state of maturation, that also depend on the precursor B-cell subset from which they derived. But, a universal definition of plasma cell subsets is not available so far. Furthermore, the ability for long-term survival and effector functions may differ, potentially in a disease-specific manner. Characterization of plasma cell subsets and their specificity in individual patients can help to choose a suitable targeting approach for either a broad or more selective plasma cell depletion. Targeting plasma cells in systemic ARDs is currently challenging because of side effects or varying depletion efficacies in the tissue. Recent developments, however, like antigen-specific targeting and CAR-T-cell therapy might open up major benefits for patients beyond current treatment options.
Collapse
Affiliation(s)
- Tobit D Steinmetz
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Gwenny M Verstappen
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jolien Suurmond
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans G M Kroese
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
39
|
Chayé MAM, Gasan TA, Ozir-Fazalalikhan A, Scheenstra MR, Zawistowska-Deniziak A, van Hengel ORJ, Gentenaar M, Manurung MD, Harvey MR, Codée JDC, Chiodo F, Heijke AM, Kalinowska A, van Diepen A, Hensbergen PJ, Yazdanbakhsh M, Guigas B, Hokke CH, Smits HH. Schistosoma mansoni egg-derived thioredoxin and Sm14 drive the development of IL-10 producing regulatory B cells. PLoS Negl Trop Dis 2023; 17:e0011344. [PMID: 37363916 DOI: 10.1371/journal.pntd.0011344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
During chronic schistosome infections, a complex regulatory network is induced to regulate the host immune system, in which IL-10-producing regulatory B (Breg) cells play a significant role. Schistosoma mansoni soluble egg antigens (SEA) are bound and internalized by B cells and induce both human and mouse IL-10 producing Breg cells. To identify Breg-inducing proteins in SEA, we fractionated SEA by size exclusion chromatography and found 6 fractions able to induce IL-10 production by B cells (out of 18) in the high, medium and low molecular weight (MW) range. The high MW fractions were rich in heavily glycosylated molecules, including multi-fucosylated proteins. Using SEA glycoproteins purified by affinity chromatography and synthetic glycans coupled to gold nanoparticles, we investigated the role of these glycan structures in inducing IL-10 production by B cells. Then, we performed proteomics analysis on active low MW fractions and identified a number of proteins with putative immunomodulatory properties, notably thioredoxin (SmTrx1) and the fatty acid binding protein Sm14. Subsequent splenic murine B cell stimulations and hock immunizations with recombinant SmTrx1 and Sm14 showed their ability to dose-dependently induce IL-10 production by B cells both in vitro and in vivo. Identification of unique Breg cells-inducing molecules may pave the way to innovative therapeutic strategies for inflammatory and auto-immune diseases.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas A Gasan
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Maaike R Scheenstra
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anna Zawistowska-Deniziak
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Oscar R J van Hengel
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Max Gentenaar
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikhael D Manurung
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael R Harvey
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Fabrizio Chiodo
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Italian National Research Council, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Anouk M Heijke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alicja Kalinowska
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- Museum and Institute of Zoology, Polish Academy of Sciences, Warsaw, Poland
| | - Angela van Diepen
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
40
|
Patel AJ, Khan N, Richter A, Naidu B, Drayson MT, Middleton GW. Deep immune B and plasma cell repertoire in non-small cell lung cancer. Front Immunol 2023; 14:1198665. [PMID: 37398676 PMCID: PMC10311499 DOI: 10.3389/fimmu.2023.1198665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/01/2023] [Indexed: 07/04/2023] Open
Abstract
Introduction B cells, which have long been thought to be minor players in the development of anti-tumor responses, have been implicated as key players in lung cancer pathogenesis and response to checkpoint blockade in patients with lung cancer. Enrichment of late-stage plasma and memory cells in the tumor microenvironment has been shown in lung cancer, with the plasma cell repertoire existing on a functional spectrum with suppressive phenotypes correlating with outcome. B cell dynamics may be influenced by the inflammatory microenvironment observed in smokers and between LUAD and LUSC. Methods Here, we show through high-dimensional deep phenotyping using mass cytometry (CyTOF), next generation RNA sequencing and multispectral immunofluorescence imaging (VECTRA Polaris) that key differences exist in the B cell repertoire between tumor and circulation in paired specimens from lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC). Results In addition to the current literature, this study provides insight into the in-depth description of the B cell contexture in Non-Small Cell Lung Cancer (NSCLC) with reference to broad clinico-pathological parameters based on our analysis of 56 patients. Our findings reinforce the phenomenon of B-cell trafficking from distant circulatory compartments into the tumour microenvironment (TME). The circulatory repertoire shows a predilection toward plasma and memory phenotypes in LUAD however no major differences exist between LUAD and LUSC at the level of the TME. B cell repertoire, amongst other factors, may be influenced by the inflammatory burden in the TME and circulation, that is, smokers and non-smokers. We have further clearly demonstrated that the plasma cell repertoire exists on a functional spectrum in lung cancer, and that the suppressive regulatory arm of this axis may play a significant role in determining postoperative outcomes as well as following checkpoint blockade. This will require further long-term functional correlation. Conclusion B and Plasma cell repertoire is very diverse and heterogeneous across different tissue compartments in lung cancer. Smoking status associates with key differences in the immune milieu and the consequent inflammatory microenvironment is likely responsible for the functional and phenotypic spectrum we have seen in the plasma cell and B cell repertoire in this condition.
Collapse
Affiliation(s)
- Akshay J. Patel
- Institute of Immunology and Immunotherapy (III), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Naeem Khan
- Institute of Immunology and Immunotherapy (III), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alex Richter
- Institute of Immunology and Immunotherapy (III), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Babu Naidu
- Institute of Inflammation and Ageing (IIA), College of Medical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Mark T. Drayson
- Institute of Immunology and Immunotherapy (III), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gary W. Middleton
- Institute of Immunology and Immunotherapy (III), College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
41
|
Uckun FM, Qazi S. Upregulated Expression of ERBB2/HER2 in Multiple Myeloma as a Predictor of Poor Survival Outcomes. Int J Mol Sci 2023; 24:9943. [PMID: 37373090 DOI: 10.3390/ijms24129943] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The main goal of the present study was to examine if the RNA-sequencing (RNAseq)-based ERBB2/HER2 expression level in malignant plasma cells from multiple myeloma (MM) patients has clinical significance for treatment outcomes and survival. We examined the relationship between the RNAseq-based ERBB2 messenger ribonucleic acid (mRNA) levels in malignant plasma cells and survival outcomes in 787 MM patients treated on contemporary standard regimens. ERBB2 was expressed at significantly higher levels than ERBB1 as well as ERBB3 across all three stages of the disease. Upregulated expression of ERBB2 mRNA in MM cells was correlated with amplified expression of mRNAs for transcription factors (TF) that recognize the ERBB2 gene promoter sites. Patients with higher levels of ERBB2 mRNA in their malignant plasma cells experienced significantly increased cancer mortality, shorter progression-free survival, and worse overall survival than other patients. The adverse impact of high ERBB2 expression on patient survival outcomes remained significant in multivariate Cox proportional hazards models that accounted for the effects of other prognostic factors. To the best of our knowledge, this is the first demonstration of an adverse prognostic impact of high-level ERBB2 expression in MM patients. Our results encourage further evaluation of the prognostic significance of high-level ERBB2 mRNA expression and the clinical potential of ERBB2-targeting therapeutics as personalized medicines to overcome cancer drug resistance in high-risk as well as relapsed/refractory MM.
Collapse
Affiliation(s)
- Fatih M Uckun
- Immuno-Oncology Program, Ares Pharmaceuticals, St. Paul, MN 55110, USA
| | - Sanjive Qazi
- Immuno-Oncology Program, Ares Pharmaceuticals, St. Paul, MN 55110, USA
| |
Collapse
|
42
|
Chen C, Ma J, Pi C, Huang W, Zhang T, Fu C, Liu W, Yang YG. PPARδ inhibition blocks the induction and function of tumor-induced IL-10 + regulatory B cells and enhances cancer immunotherapy. Cell Discov 2023; 9:54. [PMID: 37291146 DOI: 10.1038/s41421-023-00568-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
IL-10+ regulatory B cells (Bregs) play a significant role in cancer immunotherapy and their presence is an indicator of negative outcome. We found that PPARδ is significantly upregulated in tumor-induced IL-10+ Bregs with a phenotype of CD19+CD24hiIgDlo/-CD38lo or CD19+CD24hiIgDlo/-CD38hi in both mice and humans, and the level of PPARδ expression was correlated with their potential to produce IL-10 and to inhibit T cell activation. Genetic inactivation of PPARδ in B cells impaired the development and function of IL-10+ B cells, and treatment with PPARδ inhibitor diminished the induction of IL-10+ Bregs by tumor and CD40 engagement. Importantly, immunotherapy with anti-CD40 or anti-PD1 antibody achieved a markedly improved outcome in tumor-bearing mice with PPARδ deficiency in B cells or treated with PPARδ inhibitor. This study shows that PPARδ is required for the development and function of IL-10+ Bregs, providing a new and effective target for selectively blocking Bregs and improving antitumor immunotherapy.
Collapse
Affiliation(s)
- Chen Chen
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
- Centre of Oncology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jianan Ma
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Chenchen Pi
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Wei Huang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Tao Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Cong Fu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China
| | - Wentao Liu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China.
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China.
- National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin University, Changchun, Jilin, China.
- International Center of Future Science, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
43
|
Figueiredo Galvao HB, Dinh QN, Thomas JM, Wassef F, Diep H, Bobik A, Sobey CG, Drummond GR, Vinh A. Proteasome inhibition reduces plasma cell and antibody secretion, but not angiotensin II-induced hypertension. Front Cardiovasc Med 2023; 10:1184982. [PMID: 37332591 PMCID: PMC10272792 DOI: 10.3389/fcvm.2023.1184982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Depletion of mature B cells affords protection against experimental hypertension. However, whether B cell-mediated hypertension is dependent on differentiation into antibody-secreting cells (ASCs) remains unclear. Using the proteasome inhibitor, bortezomib, the present study tested the effect of ASC reduction on angiotensin II-induced hypertension. Methods Male C57BL6/J mice were infused with angiotensin II (0.7 mg/kg/day; s.c.) for 28 days via osmotic minipump to induce hypertension. Normotensive control mice received saline infusion. Bortezomib (750 μg/kg) or vehicle (0.1% DMSO) was administered (i.v.) 3 days prior to minipump implantation, and twice weekly thereafter. Systolic blood pressure was measured weekly using tail-cuff plethysmography. Spleen and bone marrow B1 (CD19+B220-), B2 (B220+CD19+) and ASCs (CD138hiSca-1+Blimp-1+) were enumerated by flow cytometry. Serum immunoglobulins were quantified using a bead-based immunoassay. Results Bortezomib treatment reduced splenic ASCs by ∼68% and ∼64% compared to vehicle treatment in normotensive (2.00 ± 0.30 vs. 0.64 ± 0.15 × 105 cells; n = 10-11) and hypertensive mice (0.52 ± 0.11 vs. 0.14 ± 0.02 × 105 cells; n = 9-11), respectively. Bone marrow ASCs were also reduced by bortezomib in both normotensive (4.75 ± 1.53 vs. 1.71 ± 0.41 × 103 cells; n = 9-11) and hypertensive mice (4.12 ± 0.82 vs. 0.89 ± 0.18 × 103 cells; n = 9-11). Consistent with ASC reductions, bortezomib reduced serum IgM and IgG2a in all mice. Despite these reductions in ASCs and antibody levels, bortezomib did not affect angiotensin II-induced hypertension over 28 days (vehicle: 182 ± 4 mmHg vs. bortezomib: 177 ± 7 mmHg; n = 9-11). Conclusion Reductions in ASCs and circulating IgG2a and IgM did not ameliorate experimental hypertension, suggesting other immunoglobulin isotypes or B cell effector functions may promote angiotensin II-induced hypertension.
Collapse
Affiliation(s)
- Hericka Bruna Figueiredo Galvao
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Quynh Nhu Dinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Jordyn M. Thomas
- Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Flavia Wassef
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Henry Diep
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Prahran, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Christopher G. Sobey
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, Australia
| | - Grant R. Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, Australia
| | - Antony Vinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
44
|
Iwata S, Hajime Sumikawa M, Tanaka Y. B cell activation via immunometabolism in systemic lupus erythematosus. Front Immunol 2023; 14:1155421. [PMID: 37256149 PMCID: PMC10225689 DOI: 10.3389/fimmu.2023.1155421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/27/2023] [Indexed: 06/01/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory autoimmune disease involving multiple organs in which B cells perform important functions such as antibody and cytokine production and antigen presentation. B cells are activated and differentiated by the primary B cell receptor, co-stimulatory molecule signals-such as CD40/CD40L-, the Toll-like receptors 7,9, and various cytokine signals. The importance of immunometabolism in the activation, differentiation, and exerting functions of B cells and other immune cells has been widely reported in recent years. However, the regulatory mechanism of immunometabolism in B cells and its involvement in SLE pathogenesis remain elusive. Similarly, the importance of the PI3K-Akt-mTOR signaling pathway, glycolytic system, and oxidative phosphorylation has been demonstrated in the mechanisms of B cell immunometabolic activation, mainly in mouse studies. However, the activation of the mTOR pathway in B cells in patients with SLE, the induction of plasmablast differentiation through metabolic and transcription factor regulation by mTOR, and the involvement of this phenomenon in SLE pathogenesis are unclear. In our studies using activated B cells derived from healthy donors and from patients with SLE, we observed that methionine, an essential amino acid, is important for mTORC1 activation. Further, we observed that splenic tyrosine kinase and mTORC1 activation synergistically induce EZH2 expression and plasmablasts by suppressing BACH2 expression through epigenomic modification. Additionally, we identified another mechanism by which the glutaminolysis-induced enhancement of mitochondrial function promotes plasmablast differentiation in SLE. In this review, we focused on the SLE exacerbation mechanisms related to the activation of immune cells-especially B cells-and immunometabolism and reported the latest findings in the field.
Collapse
Affiliation(s)
- Shigeru Iwata
- Department of Rheumatology and Clinical Immunology, Wakayama Medical University, Wakayama, Japan
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Maiko Hajime Sumikawa
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
45
|
Loisel S, Lansiaux P, Rossille D, Ménard C, Dulong J, Monvoisin C, Bescher N, Bézier I, Latour M, Cras A, Farge D, Tarte K. Regulatory B Cells Contribute to the Clinical Response After Bone Marrow-Derived Mesenchymal Stromal Cell Infusion in Patients With Systemic Sclerosis. Stem Cells Transl Med 2023; 12:194-206. [PMID: 36928395 PMCID: PMC10108721 DOI: 10.1093/stcltm/szad010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/09/2023] [Indexed: 03/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have recently emerged as an interesting therapeutic approach for patients with progressive systemic sclerosis (SSc), a rare and life-threatening orphan autoimmune disease. Whereas MSC immunomodulatory potential is considered as a central mechanism for their clinical benefit, very few data are available on the impact of MSCs on immune cell subsets in vivo. In the current extended study of a phase I/II clinical trial exploring the injection of a single dose of allogeneic bone marrow-MSCs (alloBM-MSCs) in patients with severe SSc (NCT02213705), we performed a longitudinal in-depth characterization of circulating immune cells in 19 MSC-treated patients, including 14 responders and 5 non-responders. By a combination of flow cytometry and transcriptomic analyses, we highlighted an increase in circulating CD24hiCD27posCD38lo/neg memory B cells, the main IL-10-producing regulatory B cell (Breg) subset, and an upregulation of IL10 expression in ex-vivo purified B cells, specifically in responder patients, early after the alloBM-MSC infusion. In addition, a deeper alteration of the B-cell compartment before alloBM-MSC treatment, including a higher expression of profibrotic cytokines IL6 and TGFβ by sorted B cells was associated with a non-responder clinical status. Finally, BM-MSCs were able to directly upregulate IL-10 production in activated B cells in vitro. These data suggest that cytokine-producing B cells, in particular Breg, are pivotal effectors of BM-MSC therapeutic activity in SSc. Their quantification as activity biomarkers in MSC potency assays and patient selection criteria may be considered to reach optimal clinical benefit when designing MSC-based clinical trials.
Collapse
Affiliation(s)
- Séverine Loisel
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Pauline Lansiaux
- Unité de Médecine Interne (UF 04), CRMR Maladies auto-immunes et thérapie cellulaire (MATHEC), Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, Hôpital St-Louis, Paris, France
- Université de Paris Cité, IRSL, Recherche clinique appliquée à l’hématologie, URP 3518, Paris, France
| | - Delphine Rossille
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Cédric Ménard
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Joëlle Dulong
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Céline Monvoisin
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Nadège Bescher
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Isabelle Bézier
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Maëlle Latour
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| | - Audrey Cras
- Cell Therapy Unit, Saint Louis Hospital, Assistance-Publique Hôpitaux de Paris, Paris, France
- UMR1140, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, Paris, France
| | - Dominique Farge
- Unité de Médecine Interne (UF 04), CRMR Maladies auto-immunes et thérapie cellulaire (MATHEC), Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, Hôpital St-Louis, Paris, France
- Université de Paris Cité, IRSL, Recherche clinique appliquée à l’hématologie, URP 3518, Paris, France
- Department of Medicine, McGill University, Montreal, Canada
| | - Karin Tarte
- SITI, CHU Rennes, Etablissement Français du Sang Bretagne, Rennes, France
- INSERM UMR 1236, Université Rennes, INSERM, Etablissement Français du Sang Bretagne, Rennes, France
| |
Collapse
|
46
|
Inaba A, Tuong ZK, Zhao TX, Stewart AP, Mathews R, Truman L, Sriranjan R, Kennet J, Saeb-Parsy K, Wicker L, Waldron-Lynch F, Cheriyan J, Todd JA, Mallat Z, Clatworthy MR. Low-dose IL-2 enhances the generation of IL-10-producing immunoregulatory B cells. Nat Commun 2023; 14:2071. [PMID: 37045832 PMCID: PMC10097719 DOI: 10.1038/s41467-023-37424-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
Dysfunction of interleukin-10 producing regulatory B cells has been associated with the pathogenesis of autoimmune diseases, but whether regulatory B cells can be therapeutically induced in humans is currently unknown. Here we demonstrate that a subset of activated B cells expresses CD25, and the addition of low-dose recombinant IL-2 to in vitro stimulated peripheral blood and splenic human B cells augments IL-10 secretion. Administration of low dose IL-2, aldesleukin, to patients increases IL-10-producing B cells. Single-cell RNA sequencing of circulating immune cells isolated from low dose IL2-treated patients reveals an increase in plasmablast and plasma cell populations that are enriched for a regulatory B cell gene signature. The transcriptional repressor BACH2 is significantly down-regulated in plasma cells from IL-2-treated patients, BACH2 binds to the IL-10 gene promoter, and Bach2 depletion or genetic deficiency increases B cell IL-10, implicating BACH2 suppression as an important mechanism by which IL-2 may promote an immunoregulatory phenotype in B cells.
Collapse
Affiliation(s)
- Akimichi Inaba
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Tian X Zhao
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Andrew P Stewart
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Rebeccah Mathews
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Lucy Truman
- Ear, Nose Throat Department, West Suffolk Hospital, Bury St Edmunds, UK
| | - Rouchelle Sriranjan
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jane Kennet
- Wellcome-MRC Institute of Metabolic Science-Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Linda Wicker
- Medical Sciences Division, University of Oxford, Oxford, UK
| | - Frank Waldron-Lynch
- Novartis Institutes for BioMedical Research, Autoimmunity Transplantation Inflammation, Basel, Switzerland
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, Department of Medicine, University of Cambridge, Cambridge, UK
| | - John A Todd
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
- Universite de Paris and INSERM, Paris, France
| | - Menna R Clatworthy
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK.
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
47
|
Műzes G, Sipos F. Autoimmunity and Carcinogenesis: Their Relationship under the Umbrella of Autophagy. Biomedicines 2023; 11:biomedicines11041130. [PMID: 37189748 DOI: 10.3390/biomedicines11041130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023] Open
Abstract
The immune system and autophagy share a functional relationship. Both innate and adaptive immune responses involve autophagy and, depending on the disease’s origin and pathophysiology, it may have a detrimental or positive role on autoimmune disorders. As a “double-edged sword” in tumors, autophagy can either facilitate or impede tumor growth. The autophagy regulatory network that influences tumor progression and treatment resistance is dependent on cell and tissue types and tumor stages. The connection between autoimmunity and carcinogenesis has not been sufficiently explored in past studies. As a crucial mechanism between the two phenomena, autophagy may play a substantial role, though the specifics remain unclear. Several autophagy modifiers have demonstrated beneficial effects in models of autoimmune disease, emphasizing their therapeutic potential as treatments for autoimmune disorders. The function of autophagy in the tumor microenvironment and immune cells is the subject of intensive study. The objective of this review is to investigate the role of autophagy in the simultaneous genesis of autoimmunity and malignancy, shedding light on both sides of the issue. We believe our work will assist in the organization of current understanding in the field and promote additional research on this urgent and crucial topic.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
48
|
Tarlinton DM, Ding Z, Tellier J, Nutt SL. Making sense of plasma cell heterogeneity. Curr Opin Immunol 2023; 81:102297. [PMID: 36889029 DOI: 10.1016/j.coi.2023.102297] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 03/08/2023]
Abstract
Plasma cells (PCs) are essential for the quality and longevity of protective immunity. The canonical humoral response to vaccination involves induction of germinal centers in lymph nodes followed by maintenance by bone marrow-resident PCs, although there are many variations of this theme. Recent studies have highlighted the importance of PCs in nonlymphoid organs, including the gut, central nervous system, and skin. These sites harbor PCs with distinct isotypes and possible immunoglobulin-independent functions. Indeed, bone marrow now appears unique in housing PCs derived from multiple other organs. The mechanisms through which the bone marrow maintains PC survival long-term and the impact of their diverse origins on this process remain very active areas of research.
Collapse
Affiliation(s)
- David M Tarlinton
- Department of Immunology, Monash University, Melbourne, Victoria, Australia.
| | - Zhoujie Ding
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Julie Tellier
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
49
|
Mékinian A, Quinquenel A, Belkacem KA, Kanoun F, Dondi E, Franck E, Boubaya M, Mhibik M, Baran-Marszak F, Letestu R, Ajchenbaum-Cymbalista F, Lévy V, Varin-Blank N, Le Roy C. Immuno-regulatory malignant B cells contribute to Chronic Lymphocytic Leukemia progression. Cancer Gene Ther 2023:10.1038/s41417-023-00602-5. [PMID: 36973425 DOI: 10.1038/s41417-023-00602-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 01/29/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023]
Abstract
Chronic Lymphocytic Leukemia (CLL) is a heterogeneous B cell neoplasm ranging from indolent to rapidly progressive disease. Leukemic cell subsets with regulatory properties evade immune clearance; however, the contribution of such subsets during CLL progression is not completely elucidated. Here, we report that CLL B cells crosstalk with their immune counterparts, notably by promoting the regulatory T (Treg) cell compartment and shaping several helper T (Th) subsets. Among various constitutively- and BCR/CD40-mediated factors secreted, tumour subsets co-express two important immunoregulatory cytokines, IL10 and TGFβ1, both associated with a memory B cell phenotype. Neutralizing secreted IL10 or inhibiting the TGFβ signalling pathway demonstrated that these cytokines are mainly involved in Th- and Treg differentiation/maintenance. In line with the regulatory subsets, we also demonstrated that a CLL B cell population expresses FOXP3, a marker of regulatory T cells. Analysis of IL10, TGFβ1 and FOXP3 positive subpopulations frequencies in CLL samples discriminated 2 clusters of untreated CLL patients that were significantly different in Tregs frequency and time-to-treatment. Since this distinction was pertinent to disease progression, the regulatory profiling provides a new rationale for patient stratification and sheds light on immune dysfunction in CLL.
Collapse
Affiliation(s)
- Arsène Mékinian
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | - Anne Quinquenel
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | - Koceïla Ait Belkacem
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | - Feriel Kanoun
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | - Elisabetta Dondi
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | - Emilie Franck
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | | | - Maïssa Mhibik
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
| | - Fanny Baran-Marszak
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
- Service d'Hématologie Biologique, APHP, Hôpital Avicenne, Bobigny, France
| | - Rémi Letestu
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
- Service d'Hématologie Biologique, APHP, Hôpital Avicenne, Bobigny, France
| | - Florence Ajchenbaum-Cymbalista
- INSERM, U978, Bobigny, France
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France
- Service d'Hématologie Biologique, APHP, Hôpital Avicenne, Bobigny, France
| | - Vincent Lévy
- URC, APHP, Hôpital Avicenne, Bobigny, France
- CRC, APHP, Hôpital Avicenne, Bobigny, France
| | - Nadine Varin-Blank
- INSERM, U978, Bobigny, France.
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France.
| | - Christine Le Roy
- INSERM, U978, Bobigny, France.
- Université Paris 13 dite « Sorbonne Paris Nord », UFR SMBH, Labex INFLAMEX, Bobigny, France.
| |
Collapse
|
50
|
Pioli KT, Lau KH, Pioli PD. Thymus antibody-secreting cells possess an interferon gene signature and are preferentially expanded in young female mice. iScience 2023; 26:106223. [PMID: 36890795 PMCID: PMC9986522 DOI: 10.1016/j.isci.2023.106223] [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: 06/23/2022] [Revised: 01/05/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
Antibody-secreting cells (ASCs) are key contributors to humoral immunity through immunoglobulin production and the potential to be long-lived. ASC persistence has been recognized in the autoimmune thymus (THY); however, only recently has this population been appreciated in healthy THY tissue. We showed that the young female THY was skewed toward higher production of ASCs relative to males. However, these differences disappeared with age. In both sexes, THY ASCs included Ki-67+ plasmablasts which required CD154(CD40L) signals for their propagation. Single cell RNA-sequencing revealed that THY ASCs were enriched for an interferon responsive transcriptional signature relative to those from bone marrow and spleen. Flow cytometry confirmed that THY ASCs had increased levels of Toll-like receptor 7 as well as CD69 and major histocompatibility complex class II. Overall, we identified fundamental aspects of THY ASC biology which may be leveraged for future in depth studies of this population in both health and disease.
Collapse
Affiliation(s)
- KimAnh T. Pioli
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| | - Kin H. Lau
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Peter D. Pioli
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N5E5, Canada
| |
Collapse
|