1
|
Huang W, Wang J, Liu C, Yang C, Chen Z, Ding J, Jiang W, Wang Y, Meng Y, Li L, Liu Y, Liu X, Li H, Sun B. Norepinephrine promotes activated B cells to identify and kill effector CD8 + T cells through FasL/Fas pathway in spleen mononuclear cells isolated from experimental autoimmune encephalomyelitis. Brain Behav Immun 2025; 125:294-307. [PMID: 39824471 DOI: 10.1016/j.bbi.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025] Open
Abstract
It has been reported that the nervous system can regulate immune reactions through various mechanisms. However, the role of splenic sympathetic nerve activity in the autoimmune reactions during the pathogenesis of experimental autoimmune encephalomyelitis (EAE) remained unclear. Here, we blocked the activity of the splenic sympathetic nerve and found that the number of adaptive immune cells, such as CD4+ T cells, CD8+ T cells and B cells, were upregulated. Additionally, there was an increase in the secretion of inflammatory cytokines in the spleen, and the neurological symptoms of EAE were exacerbated. In vitro experiments, we found that norepinephrine (NE), the neurotransmitter of the splenic sympathetic nerve, indirectly drove the death of effector CD8+ T cells. Furthermore, activated B cells, under the influence of NE, specifically recognized effector CD8+ T cells by upregulating MHC-I molecules and killed these cells via the FasL/Fas pathway. Our findings provide a new perspective on B cells killing effect in vitro, which was boosted by NE and demonstrate that the splenic sympathetic nerve controls the degree of autoimmune responses in EAE. This adds a new dimension to the diversity of NE's regulatory effects on adaptive immune cells and suggests a potential new therapeutic approach for autoimmune diseases.
Collapse
Affiliation(s)
- Wei Huang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Chao Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Changxin Yang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Zhengyi Chen
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Jianwen Ding
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Wenkang Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Yanping Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Yanting Meng
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Lei Li
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang, PR China
| | - Yumei Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Xijun Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Hulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China.
| | - Bo Sun
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China.
| |
Collapse
|
2
|
Motaung B, Snyders C, Malherbe S, Gutschmidt A, van Rensburg I, Loxton AG. Exogenous binding immunoglobulin protein (BiP) enhance immune regulatory phenotype in ex-vivo Mtb infected PBMCs stratified based on QuantiFERON response. Cytokine 2025; 186:156832. [PMID: 39671882 DOI: 10.1016/j.cyto.2024.156832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Even though anti-tuberculosis (TB) treatment is readily available, Mycobacterium tuberculosis (Mtb) infection continues to be a global threat with a high death rate recorded from a single infectious agent. This highlights the significance of developing new strategies to curb the growing Mtb infection cases. Host-directed therapies (HDT) offer a promising approach that includes both drug discovery and drug repurposing, aimed at identifying host targets and promoting immune cell populations that can lead to better infection outcomes. In this context, we investigated the potential of exogenous Binding Immunoglobulin Protein (BiP) to induce such changes ex-vivo using PBMCs from healthy (QFN-) and Mtb exposed (QFN+) individuals. We analysed cell surface expression and cytokine profiles across eight different stimulation conditions including human full-length BiP protein (20 μg/ml), TLR-9a (0.5 μM), BiP/TLR-9a combination, isoniazid (1 μM), H37Rv (MOI: 1: 10), and pooled bronchoalveolar lavage (BAL) samples collected at TB diagnosis (TBdx) and at month 6 (M6) of anti-TB treatment. Our results revealed that BiP-stimulated PBMCs showed a significant reduction of interleukin (IL)-10 secretion, along with increased IL-4, IL-5, IL-13, and soluble Fas-L (sFasL) secretion. We also observed that BiP stimulation enhanced the expression of membrane bound Fas-L (CD178) and IL5Ra (CD125) in B-cells isolated from both QFN- and QFN+ groups. Additionally, BiP exhibited a synergistic effect with TLR-9a, further boosting this co-expression. Moreover, we observed that BiP induced IL5Ra expression in both CD3+CD5lo and CD3+CD5hi T-cell populations. This study explores the effects of exogenous BiP on cell functionality and provides valuable insights into its potential to modulate host cell responses, which could be explored as a host-directed therapy for TB in the future.
Collapse
Affiliation(s)
- Bongani Motaung
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Candice Snyders
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Stephanus Malherbe
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andrea Gutschmidt
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ilana van Rensburg
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research; South African Medical Research Council Centre for Tuberculosis Research; Division of Immunology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre G Loxton
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
3
|
Bryushkova EA, Mushenkova NV, Turchaninova MA, Lukyanov DK, Chudakov DM, Serebrovskaya EO. B cell clonality in cancer. Semin Immunol 2024; 72:101874. [PMID: 38508089 DOI: 10.1016/j.smim.2024.101874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 03/22/2024]
Abstract
Carcinogenesis in the process of long-term co-evolution of tumor cells and immune environment essentially becomes possible due to incorrect decisions made, remembered, and reproduced by the immune system at the level of clonal populations of antigen-specific T- and B-lymphocytes. Tumor-immunity interaction determines the nature of such errors and, consequently, delineates the possible ways of successful immunotherapeutic intervention. It is generally recognized that tumor-infiltrating B cells (TIL-B) can play both pro-tumor and anti-tumor roles. However, the exact mechanisms that determine the contribution of clonal B cell lineages with different specificities and functions remain largely unclear. This is due to the variability of cancer types, the molecular heterogeneity of tumor cells, and, to a large extent, the individual pattern of each immune response. Further progress requires detailed investigation of the functional properties and phenotypes of clonally heterogeneous B cells in relation to their antigenic specificities, which determine the functionality of both effector B lymphocytes and immunoglobulins produced in the tumor environment. Based on a real understanding of the role of clonal antigen-specific populations of B lymphocytes in the tumor microenvironment, we need to learn how to develop new methods of targeted immunotherapy, as well as adapt existing treatment options to the specific needs of different patients and patient subgroups. In this review, we will cover B cells functional diversity and their multifaceted roles in the tumor environment.
Collapse
Affiliation(s)
- E A Bryushkova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Department of Molecular Biology, Lomonosov Moscow State University, Moscow, Russia
| | - N V Mushenkova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Unicorn Capital Partners, Moscow, Russia
| | - M A Turchaninova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia
| | - D K Lukyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - D M Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia; Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
| | - E O Serebrovskaya
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Moscow, Russia; Current position: Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| |
Collapse
|
4
|
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
|
5
|
Matsumura Y, Watanabe R, Fujimoto M. Suppressive mechanisms of regulatory B cells in mice and humans. Int Immunol 2022; 35:55-65. [PMID: 36153768 PMCID: PMC9918854 DOI: 10.1093/intimm/dxac048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/23/2022] [Indexed: 11/14/2022] Open
Abstract
B cells include immune-suppressive fractions, called regulatory B cells (Bregs), which regulate inflammation primarily through an interleukin 10 (IL-10)-mediated inhibitory mechanism. Several B-cell fractions have been reported as IL-10-producing Bregs in murine disease models and human inflammatory responses including autoimmune diseases, infectious diseases, cancer and organ-transplant rejection. Although the suppressive functions of Bregs have been explored through the hallmark molecule IL-10, inhibitory cytokines and membrane-binding molecules other than IL-10 have also been demonstrated to contribute to Breg activities. Transcription factors and surface antigens that are characteristically expressed in Bregs are also being elucidated. Nevertheless, defining Bregs is still challenging because their active periods and differentiation stages vary among disease models. The identity of the diverse Breg fractions is also under debate. In the first place, since regulatory functions of Bregs are mostly evaluated by ex vivo stimulation, the actual in vivo phenotypes and functions may not be reflected by the ex vivo observations. In this article, we provide a historical overview of studies that established the characteristics of Bregs and review the various suppressive mechanisms that have been reported to be used by Bregs in murine and human disease conditions. We are only part-way through but the common phenotypes and functions of Bregs are still emerging.
Collapse
Affiliation(s)
- Yutaka Matsumura
- Department of Dermatology, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka, 565-0871, Japan
| | - Rei Watanabe
- Department of Dermatology, Graduate School of Medicine, Faculty of Medicine, Osaka University, Osaka, 565-0871, Japan,Department of Integrative Medicine for Allergic and Immunological Diseases, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, 565-0871, Japan
| | | |
Collapse
|
6
|
Sakowska J, Arcimowicz Ł, Jankowiak M, Papak I, Markiewicz A, Dziubek K, Kurkowiak M, Kote S, Kaźmierczak-Siedlecka K, Połom K, Marek-Trzonkowska N, Trzonkowski P. Autoimmunity and Cancer-Two Sides of the Same Coin. Front Immunol 2022; 13:793234. [PMID: 35634292 PMCID: PMC9140757 DOI: 10.3389/fimmu.2022.793234] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Autoimmune disease results from the immune response against self-antigens, while cancer develops when the immune system does not respond to malignant cells. Thus, for years, autoimmunity and cancer have been considered as two separate fields of research that do not have a lot in common. However, the discovery of immune checkpoints and the development of anti-cancer drugs targeting PD-1 (programmed cell death receptor 1) and CTLA-4 (cytotoxic T lymphocyte antigen 4) pathways proved that studying autoimmune diseases can be extremely helpful in the development of novel anti-cancer drugs. Therefore, autoimmunity and cancer seem to be just two sides of the same coin. In the current review, we broadly discuss how various regulatory cell populations, effector molecules, genetic predisposition, and environmental factors contribute to the loss of self-tolerance in autoimmunity or tolerance induction to cancer. With the current paper, we also aim to convince the readers that the pathways involved in cancer and autoimmune disease development consist of similar molecular players working in opposite directions. Therefore, a deep understanding of the two sides of immune tolerance is crucial for the proper designing of novel and selective immunotherapies.
Collapse
Affiliation(s)
- Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Łukasz Arcimowicz
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Martyna Jankowiak
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ines Papak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Sachin Kote
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | | | - Karol Połom
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
7
|
Honke N, Lowin T, Opgenoorth B, Shaabani N, Lautwein A, Teijaro JR, Schneider M, Pongratz G. Endogenously produced catecholamines improve the regulatory function of TLR9-activated B cells. PLoS Biol 2022; 20:e3001513. [PMID: 35073310 PMCID: PMC8786184 DOI: 10.1371/journal.pbio.3001513] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023] Open
Abstract
The sympathetic nervous system (SNS) contributes to immune balance by promoting anti-inflammatory B cells. However, whether B cells possess a self-regulating mechanism by which they modulate regulatory B cell (Breg) function is not well understood. In this study, we investigated the ability of B cells to synthesize their own catecholamines upon stimulation with different B cell activators and found that expression of the enzyme tyrosine hydroxylase (TH), required to generate catecholamines, is up-regulated by Toll-like receptor (TLR)9. This TLR9-dependent expression of TH correlated with up-regulation of adrenergic receptors (ADRs), enhanced interleukin (IL)-10 production, and overexpression of the co-inhibitory ligands programmed death ligand 1 (PD-L1) and Fas ligand (FasL). Moreover, concomitant stimulation of ß1-3-ADRs together with a B cell receptor (BCR)/TLR9 stimulus clearly enhances the anti-inflammatory potential of Bregs to suppress CD4 T cells, a crucial population in the pathogenesis of autoimmune diseases, like rheumatoid arthritis (RA). Furthermore, TH up-regulation was also demonstrated in B cells during the course of collagen-induced arthritis (CIA), a mouse model for the investigation of RA. In conclusion, our data show that B cells possess an autonomous mechanism to modulate their regulatory function in an autocrine and/or paracrine manner. These findings help to better understand the function of B cells in the regulation of autoimmune diseases and the interplay of SNS. The sympathetic nervous system produces neurotransmitters such as catecholamines which contribute to immune balance by promoting anti-inflammatory B cells. This study shows that mouse B cells can themselves synthesize, sense, and transport catecholamines, which in turn modulate regulatory B cell function in an autocrine and/or paracrine manner to suppress T cell proliferation.
Collapse
Affiliation(s)
- Nadine Honke
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
- * E-mail: (NH); (GP)
| | - Torsten Lowin
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - Birgit Opgenoorth
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - Namir Shaabani
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Alexander Lautwein
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - John R. Teijaro
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Matthias Schneider
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
| | - Georg Pongratz
- Department of Rheumatology, Hiller Research Center Rheumatology, University Hospital Düsseldorf, Germany
- * E-mail: (NH); (GP)
| |
Collapse
|
8
|
Rosser EC, Mauri C. The emerging field of regulatory B cell immunometabolism. Cell Metab 2021; 33:1088-1097. [PMID: 34077716 DOI: 10.1016/j.cmet.2021.05.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 03/31/2021] [Accepted: 05/05/2021] [Indexed: 01/02/2023]
Abstract
B cells are well known as critical mediators of humoral immune responses via the production of antibodies. However, numerous studies have also identified populations of B cells that are characterized by their anti-inflammatory properties. These "regulatory B cells" restrain excessive inflammatory responses in a wide range of health conditions. A significant knowledge gap remains concerning the nature of the signals that determine whether a B cell exerts a pro-inflammatory or anti-inflammatory function. In this perspective, we explore the concept that in addition to the cytokine microenvironment, intracellular and extracellular metabolic signals play a pivotal role in controlling the balance between regulatory and antibody-producing B cell subsets. Determining the metabolites and tissue-specific signals that influence B cell fate could establish novel therapeutic targets for the treatment of diseases where abnormal B cell responses contribute to pathogenesis.
Collapse
Affiliation(s)
- Elizabeth C Rosser
- Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London Hospital and Great Ormond Street Hospital, University College London, London, UK; Centre for Rheumatology Research, Division of Medicine, University College London, London, UK.
| | - Claudia Mauri
- Division of Infection, Immunity and Transplantation, University College London, London, UK
| |
Collapse
|
9
|
Catalán D, Mansilla MA, Ferrier A, Soto L, Oleinika K, Aguillón JC, Aravena O. Immunosuppressive Mechanisms of Regulatory B Cells. Front Immunol 2021; 12:611795. [PMID: 33995344 PMCID: PMC8118522 DOI: 10.3389/fimmu.2021.611795] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
Regulatory B cells (Bregs) is a term that encompasses all B cells that act to suppress immune responses. Bregs contribute to the maintenance of tolerance, limiting ongoing immune responses and reestablishing immune homeostasis. The important role of Bregs in restraining the pathology associated with exacerbated inflammatory responses in autoimmunity and graft rejection has been consistently demonstrated, while more recent studies have suggested a role for this population in other immune-related conditions, such as infections, allergy, cancer, and chronic metabolic diseases. Initial studies identified IL-10 as the hallmark of Breg function; nevertheless, the past decade has seen the discovery of other molecules utilized by human and murine B cells to regulate immune responses. This new arsenal includes other anti-inflammatory cytokines such IL-35 and TGF-β, as well as cell surface proteins like CD1d and PD-L1. In this review, we examine the main suppressive mechanisms employed by these novel Breg populations. We also discuss recent evidence that helps to unravel previously unknown aspects of the phenotype, development, activation, and function of IL-10-producing Bregs, incorporating an overview on those questions that remain obscure.
Collapse
Affiliation(s)
- Diego Catalán
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Miguel Andrés Mansilla
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Ashley Ferrier
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Instituto Milenio en Inmunología e Inmunoterapia, Santiago, Chile
| | - Lilian Soto
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile.,Unidad de Dolor, Hospital Clínico, Universidad de Chile (HCUCH), Santiago, Chile
| | | | - Juan Carlos Aguillón
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| | - Octavio Aravena
- Programa Disciplinario de Inmunología, Facultad de Medicina, Instituto de Ciencias Biomédicas (ICBM), Universidad de Chile, Santiago, Chile
| |
Collapse
|
10
|
Lundy SK, Taitano SH, van der Vlugt LEPM. Characterization and Activation of Fas Ligand-Producing Mouse B Cells and Their Killer Exosomes. Methods Mol Biol 2021; 2270:149-178. [PMID: 33479898 DOI: 10.1007/978-1-0716-1237-8_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
B lymphocytes make several contributions to immune regulation including production of antibodies with regulatory properties, release of immune suppressive cytokines, and expression of death-inducing ligands. A role for Fas ligand (FasL)-expressing "killer" B cells in regulating T helper (TH) cell survival and chronic inflammation has been demonstrated in animal models of schistosome worm and other infections, asthma, autoimmune arthritis, and type 1 diabetes. FasL+ B cells were also capable of inducing immune tolerance in a male-to-female transplantation model. Interestingly, populations of B cells found in the spleen and lungs of naïve mice constitutively expresses FasL and have potent killer function against TH cells that is antigen-specific and FasL-dependent. Epstein-Barr virus-transformed human B cells constitutively express FasL and package it into exosomes that co-express MHC Class II molecules and have killer function against antigen-specific TH cells. FasL+ exosomes with markers of B-cell lineage are abundant in the spleen of naïve mice. Killer B cells therefore represent a novel target for immune modulation in many disease settings. Our laboratory has published methods of characterizing FasL+ B cells and inducing their proliferation in vitro. This updated chapter will describe methods of identifying and expanding killer B cells from mice, detecting FasL expression in B cells, extracting FasL+ exosomes from spleen and culture supernatants, and performing functional killing assays against antigen-specific TH cells.
Collapse
Affiliation(s)
- Steven K Lundy
- Graduate Program in Immunology, Program in Biomedical Sciences and Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Sophina H Taitano
- Graduate Program in Immunology, Program in Biomedical Sciences and Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Luciën E P M van der Vlugt
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Ma S, Satitsuksanoa P, Jansen K, Cevhertas L, van de Veen W, Akdis M. B regulatory cells in allergy. Immunol Rev 2020; 299:10-30. [PMID: 33345311 DOI: 10.1111/imr.12937] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022]
Abstract
B cells have classically been recognized for their unique and indispensable role in the production of antibodies. Their potential as immunoregulatory cells with anti-inflammatory functions has received increasing attention during the last two decades. Herein, we highlight pioneering studies in the field of regulatory B cell (Breg) research. We will review the literature on Bregs with a particular focus on their role in the regulation of allergic inflammation.
Collapse
Affiliation(s)
- Siyuan Ma
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | | | - Kirstin Jansen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Department of Medical Immunology, Institute of Health Sciences, Bursa Uludag University, Bursa, Turkey
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
12
|
Dubois F, Limou S, Chesneau M, Degauque N, Brouard S, Danger R. Transcriptional meta-analysis of regulatory B cells. Eur J Immunol 2020; 50:1757-1769. [PMID: 32529638 DOI: 10.1002/eji.201948489] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/01/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022]
Abstract
Regulatory B cells (Bregs) have the ability to regulate inflammation in various pathological situations, making them key players in immune regulation. Several mechanisms have been described and we recently identified a GZMB expressing Breg population in kidney transplanted patients who tolerate a kidney graft. To further investigate their biology and mechanisms, we conducted a transcriptomic analysis by RNAseq of these cells and we performed the first weighted meta-analysis of publicly available transcriptomic data from published Breg studies both in humans and mice. We identified two distinct and unique transcriptional signatures of 126 and 93 genes, respectively, associated with these Bregs. While we highlighted genes coding for proteins with potent involvement in regulatory functions, proliferation, and coding for transcription factors, the comparison between humans and mice did not allow identifying a common pattern. Thus, our results suggest distinct species-restricted Breg transcriptional signatures in humans and mice.
Collapse
Affiliation(s)
- Florian Dubois
- Inserm, CHU Nantes, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France
| | - Sophie Limou
- Inserm, CHU Nantes, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Ecole Centrale de Nantes, Computer Sciences and Mathematics department, Nantes, France
| | - Mélanie Chesneau
- Inserm, CHU Nantes, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France
| | - Nicolas Degauque
- Inserm, CHU Nantes, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France
| | - Sophie Brouard
- Inserm, CHU Nantes, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France.,Centre d'Investigation Clinique en Biothérapie, Centre de ressources biologiques (CRB), Nantes, France
| | - Richard Danger
- Inserm, CHU Nantes, Université de Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,Labex IGO, Nantes, France
| |
Collapse
|
13
|
Piper CJM, Rosser EC, Oleinika K, Nistala K, Krausgruber T, Rendeiro AF, Banos A, Drozdov I, Villa M, Thomson S, Xanthou G, Bock C, Stockinger B, Mauri C. Aryl Hydrocarbon Receptor Contributes to the Transcriptional Program of IL-10-Producing Regulatory B Cells. Cell Rep 2019; 29:1878-1892.e7. [PMID: 31722204 PMCID: PMC6856759 DOI: 10.1016/j.celrep.2019.10.018] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/23/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022] Open
Abstract
Regulatory B cells (Bregs) play a critical role in the control of autoimmunity and inflammation. IL-10 production is the hallmark for the identification of Bregs. However, the molecular determinants that regulate the transcription of IL-10 and control the Breg developmental program remain unknown. Here, we demonstrate that aryl hydrocarbon receptor (AhR) regulates the differentiation and function of IL-10-producing CD19+CD21hiCD24hiBregs and limits their differentiation into B cells that contribute to inflammation. Chromatin profiling and transcriptome analyses show that loss of AhR in B cells reduces expression of IL-10 by skewing the differentiation of CD19+CD21hiCD24hiB cells into a pro-inflammatory program, under Breg-inducing conditions. B cell AhR-deficient mice develop exacerbated arthritis, show significant reductions in IL-10-producing Bregs and regulatory T cells, and show an increase in T helper (Th) 1 and Th17 cells compared with B cell AhR-sufficient mice. Thus, we identify AhR as a relevant contributor to the transcriptional regulation of Breg differentiation.
Collapse
Affiliation(s)
- Christopher J M Piper
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Elizabeth C Rosser
- Centre for Rheumatology, Division of Medicine, University College London, London, UK; University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; Versus Arthritis Centre for Adolescent Rheumatology at University College London, University College London Hospitals and Great Ormond Street Hospital, London, UK
| | - Kristine Oleinika
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Kiran Nistala
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - André F Rendeiro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Aggelos Banos
- Laboratory of Inflammation and Autoimmunity, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | | | - Matteo Villa
- The Francis Crick Institute, London, NW1 1AT, UK
| | - Scott Thomson
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Georgina Xanthou
- Cellular Immunology Lab, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Max Planck Institute for Informatics, Saarland Informatics Campus, Saarbrücken, Germany
| | | | - Claudia Mauri
- Centre for Rheumatology, Division of Medicine, University College London, London, UK.
| |
Collapse
|
14
|
Tao L, Wang Y, Xu J, Su J, Yang Q, Deng W, Zou B, Tan Y, Ding Z, Li X. IL-10-producing regulatory B cells exhibit functional defects and play a protective role in severe endotoxic shock. Pharmacol Res 2019; 148:104457. [PMID: 31536782 DOI: 10.1016/j.phrs.2019.104457] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/20/2019] [Accepted: 09/15/2019] [Indexed: 02/06/2023]
Abstract
Dysregulated host immune homeostasis in sepsis is life-threatening even after a successfully treated bacterial infection. Lipopolysaccharide (LPS) is an endotoxin that is a major contributor to the aberrant immune responses and endotoxic shock in gram-negative bacterial sepsis. However, the current knowledge of the role of B cells in endotoxic shock is limited. Here, we report that CD1d expression in B cells and the percentage of CD5+CD1dhi regulatory B (Breg) cells decreased in a mouse model of endotoxic shock. Interestingly, IL-10 but not FasL expression in CD5+CD1dhi Breg cells in response to endotoxin was dramatically reduced in severe septic shock mice, and the regulatory function of CD5+CD1dhi Breg cells in vitro to control the Th1 response was also diminished. Adoptive transfer of CD5+CD1dhi Breg cells from healthy WT mice but not IL-10 deficient mice downregulated the IFN-γ secretion in CD4+ T cells and conferred protection against severe endotoxic shock in vivo. Our findings demonstrate the change and notable therapeutic potential of IL-10-producing Breg cells in endotoxic shock.
Collapse
Affiliation(s)
- Lei Tao
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd., Guangzhou, China
| | - Yiyuan Wang
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jialan Xu
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jianbing Su
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qin Yang
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wende Deng
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Binhua Zou
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yanhui Tan
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zongbao Ding
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xiaojuan Li
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
15
|
Karim MR, Wang YF. Phenotypic identification of CD19 +CD5 +CD1d + regulatory B cells that produce interleukin 10 and transforming growth factor β 1 in human peripheral blood. Arch Med Sci 2019; 15:1176-1183. [PMID: 31572462 PMCID: PMC6764295 DOI: 10.5114/aoms.2018.77772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Regulatory B cells (Bregs), a novel subpopulation of B cells, are a significant area of research due to their immune regulatory function in the immunological response. Bregs have been reported to regulate acute inflammation and immunity through the production of anti-inflammatory cytokines. MATERIAL AND METHODS A B cell subpopulation was identified using flow cytometric analysis in two different processes: 1) after preparation and storage of peripheral blood mononuclear cells (PBMCs) using Ficoll density gradient centrifugation from a human blood sample, 2) followed by isolation and storage of B cells through magnetic separation using a B cell isolation kit and MS column. ELISA assays were performed to observe the cytokine production of interkleukin 10 (IL-10) and transforming growth factor β1 (TGF-β1) by this novel B cell subpopulation. RESULTS Double positive staining of CD5+CD1d+ Bregs represents (19.27 ±1.52) from PBMCs, (33.32 ±2.95) from B cells accordingly (n = 40). Through ELISA assays, it has been found that B cell subpopulation produces IL-10 (0.56 ±0.08) and TGF-β1 (0.90 ±0.12) (n = 40). CONCLUSIONS These methods should be able to facilitate progress in research on Bregs through the following steps: 1) the regulatory role may be observed in comparison with particular autoimmune diseases, inflammation, cancer, and immunologic responses to find out whether Breg alteration and/or cytokine production is altered as well in these disorders or conditions. 2) If the alteration of Bregs and cytokine production is significant along with the clinical correlation, a further in vitro study can be initiated with exposure of certain drugs to overcome the alteration of the cytokine production; then, an in vivo study can be initiated.
Collapse
Affiliation(s)
- Md Rezaul Karim
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China
- Biomedical Research Institute of Hubei University of Medicine, Shiyan, China
| | - Yun-Fu Wang
- Department of Neurology, Taihe Hospital of Hubei University of Medicine, Shiyan, China
- Biomedical Research Institute of Hubei University of Medicine, Shiyan, China
| |
Collapse
|
16
|
Su J, Wang K, Zhou X, Wang Y, Xu J, Tao L, Zeng X, Chen N, Bai X, Li X. B-cell-specific-peroxisome proliferator-activated receptor γ deficiency augments contact hypersensitivity with impaired regulatory B cells. Immunology 2018; 156:282-296. [PMID: 30471095 DOI: 10.1111/imm.13027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/13/2022] Open
Abstract
Nuclear receptor peroxisome proliferator-activated receptor γ (PPAR-γ) activation can prevent immunoinflammatory disorders and diabetes. B cells play protective roles during inflammation as well. However, the roles of endogenous PPAR-γ in the regulatory properties of B cells to relieve inflammation remain unknown. Here, we developed B-cell-specific PPAR-γ knockout (B-PPAR-γ-/- ) mice and found that the conditional deletion of PPAR-γ in B cells resulted in exaggerated contact hypersensitivity (CHS). Meanwhile, interferon-γ (IFN-γ) of CD4+ CD8+ T cells was up-regulated in B-PPAR-γ-/- mice in CHS. This showed that the regulatory function of B cells in B-PPAR-γ-/- mice declined in vivo. Whereas splenic CD5+ CD1dhi regulatory B-cell numbers and peripheral regulatory T-cell numbers were not changed in naive B-PPAR-γ-/- mice. Loss of PPAR-γ in B cells also did not affect either CD86 or FasL expression in splenic CD5+ CD1dhi regulatory B cells after activation. Notably, interleukin-10 (IL-10) production in CD5+ CD1dhi regulatory B cells reduced in B-PPAR-γ-deficient mice. In addition, functional IL-10-producing CD5+ CD1dhi regulatory B cells decreased in B-PPAR-γ-/- mice in the CHS model. These findings were in accordance with augmented CHS. The current work indicated the involvement of endogenous PPAR-γ in the regulatory function of B cells by disturbing the expansion of IL-10-positive regulatory B cells.
Collapse
Affiliation(s)
- Jianbing Su
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Keng Wang
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xuan Zhou
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yiyuan Wang
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jialan Xu
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Lei Tao
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiangzhou Zeng
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Nana Chen
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaojuan Li
- Laboratory of Anti-inflammatory and Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Vonberg AD, Acevedo-Calado M, Cox AR, Pietropaolo SL, Gianani R, Lundy SK, Pietropaolo M. CD19+IgM+ cells demonstrate enhanced therapeutic efficacy in type 1 diabetes mellitus. JCI Insight 2018; 3:99860. [PMID: 30518692 DOI: 10.1172/jci.insight.99860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
We describe a protective effect on autoimmune diabetes and reduced destructive insulitis in NOD.scid recipients following splenocyte injections from diabetic NOD donors and sorted CD19+ cells compared with NOD.scid recipients receiving splenocytes alone. This protective effect was age specific (only CD19+ cells from young NOD donors exerted this effect; P < 0.001). We found that the CD19+IgM+ cell is the primary subpopulation of B cells that delayed transfer of diabetes mediated by diabetogenic T cells from NOD mice (P = 0.002). Removal of IgM+ cells from the CD19+ pool did not result in protection. Notably, protection conferred by CD19+IgM+ cotransfers were not dependent on the presence of Tregs, as their depletion did not affect their ability to delay onset of diabetes. Blockade of IL-10 with neutralizing antibodies at the time of CD19+ cell cotransfers also abrogated the therapeutic effect, suggesting that IL-10 secretion was an important component of protection. These results were strengthened by ex vivo incubation of CD19+ cells with IL-5, resulting in enhanced proliferation and IL-10 production and equivalently delayed diabetes progression (P = 0.0005). The potential to expand CD19+IgM+ cells, especially in response to IL-5 stimulation or by pharmacologic agents, may be a new therapeutic option for type 1 diabetes.
Collapse
Affiliation(s)
- Andrew D Vonberg
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Maria Acevedo-Calado
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Aaron R Cox
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Susan L Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Roberto Gianani
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| | - Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Massimo Pietropaolo
- Diabetes Research Center, Division of Diabetes, Endocrinology and Metabolism Department of Medicine, and
| |
Collapse
|
18
|
Zhang H, Wang Z, Liu R, Qian T, Liu J, Wang L, Chu Y. Reactive oxygen species stimulated pulmonary epithelial cells mediate the alveolar recruitment of FasL
+
killer B cells in LPS‐induced acute lung injuries. J Leukoc Biol 2018; 104:1187-1198. [DOI: 10.1002/jlb.3a0218-075r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 07/19/2018] [Accepted: 08/14/2018] [Indexed: 11/11/2022] Open
Affiliation(s)
- Hushan Zhang
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Zhiming Wang
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Ronghua Liu
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Tingting Qian
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Jiajing Liu
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
| | - Luman Wang
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
- Biotherapy Research CenterFudan University Shanghai China
| | - Yiwei Chu
- Department of ImmunologySchool of Basic Medical SciencesInstitute of Biomedical SciencesFudan University Shanghai China
- Biotherapy Research CenterFudan University Shanghai China
| |
Collapse
|
19
|
Taitano SH, van der Vlugt LEPM, Shea MM, Yang J, Lukacs NW, Lundy SK. Differential Influence on Regulatory B Cells by T H2 Cytokines Affects Protection in Allergic Airway Disease. THE JOURNAL OF IMMUNOLOGY 2018; 201:1865-1874. [PMID: 30127086 DOI: 10.4049/jimmunol.1800206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/23/2018] [Indexed: 11/19/2022]
Abstract
The role of regulatory B cells (Bregs) in modulating immune responses and maintaining tolerance are well established. However, how cytokines present during immune responses affect Breg growth and function are not as well defined. Previously, our laboratory reported IL-5- and mCD40L-expressing fibroblast (mCD40L-Fb) stimulation induced IL-10 production from murine B cells. The current study investigated the phenotype and functional relevance of IL-10- producing B cells from this culture. We found IL-5/mCD40L-Fb stimulation induced IL-10 production exclusively from CD5+ splenic B cells of naive mice. After stimulation, the resulting IL-10+ B cells displayed markers of multiple reported Breg phenotypes. Interestingly, when investigating effects of IL-4 (a critical TH2 cytokine) on IL-5/mCD40L-Fb-induced IL-10 production, we found IL-4 inhibited IL-10 production in a STAT6-dependent manner. Upon adoptive transfer, CD5+ B cells previously stimulated with IL-5/mCD40L-Fb were able to reduce development of OVA-induced allergic airway disease in mice. Using B cells from IL-10 mutant mice differentiated by IL-5/mCD40L-Fb, we found protection from allergic airway disease development was dependent on the IL-10 production from the transferred B cells. Bregs have been shown to play crucial roles in the immune tolerance network, and understanding stimuli that modulate their growth and function may be key in development of future treatments for diseases of immune dysregulation.
Collapse
Affiliation(s)
- Sophina H Taitano
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Luciën E P M van der Vlugt
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Molly M Shea
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jennifer Yang
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Nicholas W Lukacs
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and.,Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; .,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109; and
| |
Collapse
|
20
|
Abstract
PURPOSE OF REVIEW Allergen immunotherapy (AIT) is currently the only curative treatment available for allergic diseases, and has been used in clinical practice for over a century. Induction and maintenance of immune tolerance to nonhazardous environmental and self-antigens is essential to maintain homeostasis and prevent chronic inflammation. Regulatory B (BREG) cells are immunoregulatory cells that protect against chronic inflammatory responses primarily through production of anti-inflammatory cytokines such as IL-10, transforming growth factor-β, and IL-35. The importance of BREG cells has been extensively demonstrated in the context of autoimmune diseases. Data showing their role in the regulation of allergic responses are slowly accumulating. This review summarizes recent findings relevant to the topic of BREG cells and their potential role in AIT. RECENT FINDINGS BREG cells support AIT in models of allergic airway inflammation and intestinal inflammation through induction of regulatory T (TREG) cells. In humans BREG frequency increases during venom immunotherapy while the phenotype of allergen-specific B cells changes. Mechanisms of BREG-mediated tolerance to allergens include IL-10-mediated suppression of effector T cell, including TH2 responses, induction of TREG cells, IL-10-mediated inhibition of Dendritic cell maturation, modulation of T follicular helper responses, and production of anti-inflammatory IgG4 antibodies. SUMMARY Current evidence supports a potential role for BREG cells in induction and maintenance of allergen tolerance during AIT. A better understanding of the role of B cells and BREG cells in AIT could open potential new windows for developing targeted therapies specifically focused on promoting BREG responses during AIT.
Collapse
|
21
|
van Rensburg IC, Wagman C, Stanley K, Beltran C, Ronacher K, Walzl G, Loxton AG. Successful TB treatment induces B-cells expressing FASL and IL5RA mRNA. Oncotarget 2018; 8:2037-2043. [PMID: 27682872 PMCID: PMC5356777 DOI: 10.18632/oncotarget.12184] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022] Open
Abstract
Activated B-cells increase T-cell behaviour during autoimmune disease and other infections by means of cytokine production and antigen-presentation. Functional studies in experimental autoimmune encephalomyelitis (EAE) indicate that B-cell deficiencies, and a lack of IL10 and IL35 leads to a poor prognosis. We hypothesised that B-cells play a role during tuberculosis. We evaluated B-cell mRNA expression using real-time PCR from healthy community controls, individuals with other lung diseases and newly diagnosed untreated pulmonary TB patients at three different time points (diagnosis, month 2 and 6 of treatment). We show that FASLG, IL5RA, CD38 and IL4 expression was lower in B-cells from TB cases compared to healthy controls. The changes in expression levels of CD38 may be due to a reduced activation of B-cells from TB cases at diagnosis. By month 2 of treatment, there was a significant increase in the expression of APRIL and IL5RA in TB cases. Furthermore, after 6 months of treatment, APRIL, FASLG, IL5RA and CD19 were upregulated in B-cells from TB cases. The increase in the expression of APRIL and CD19 suggests that there may be restored activation of B-cells following anti-TB treatment. The upregulation of FASLG and IL5RA indicates that B-cells expressing regulatory genes may play an important role in the protective immunity against M.tb infection. Our results show that increased activation of B-cells is present following successful TB treatment, and that the expression of FASLG and IL5RA could potentially be utilised as a signature to monitor treatment response.
Collapse
Affiliation(s)
- Ilana C van Rensburg
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Chandre Wagman
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Kim Stanley
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Caroline Beltran
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Katharina Ronacher
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andre G Loxton
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
22
|
Killer (FASL regulatory) B cells are present during latent TB and are induced by BCG stimulation in participants with and without latent tuberculosis. Tuberculosis (Edinb) 2018. [DOI: 10.1016/j.tube.2017.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
23
|
Hirose T, Tanaka Y, Tanaka A, Sakai H, Sasaki Y, Shinohara N, Ohdan H. PD-L1/PD-L2-expressing B-1 cells inhibit alloreactive T cells in mice. PLoS One 2017; 12:e0178765. [PMID: 28570665 PMCID: PMC5453578 DOI: 10.1371/journal.pone.0178765] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 05/18/2017] [Indexed: 11/19/2022] Open
Abstract
B cells constitute a complex system of antigen-presenting cells (APCs) and exist as distinct subsets that differ in their lineage affiliation, surface molecule expression, and biological function, thus potentially regulating the immune response. In this study, we investigated the immune-regulatory roles of murine B cell subsets as regulatory APCs targeting alloreactive T cells. Either splenic B cells, peritoneal cavity (PerC) B cells, or non-B cells from Balb/c mice were intravenously injected into B6 mice. Serum levels of anti-Balb/c antibodies in the recipients of PerC B cells were significantly lower than those in the recipients of splenic B cells and PerC non-B cells, as determined over a 4-week period after the injection. Mixed-lymphocyte reaction (MLR) assays using splenocytes from the B6 mice at 2 weeks after the injection revealed the significantly reduced anti-Balb/c T cell-responses in the recipients of PerC B cells, as compared to those in the recipients of splenic B cells or untreated control mice. Since PerC B cells contained MHC class II+ CD80+ CD86+ PD-L1+ PD-L2+ cells among the CD5+ B-1a cell subset, PerC B cells from Balb/c mice were pre-incubated with anti-PD-L1/PD-L2 mAbs prior to injection. This treatment abrogated their immune-regulatory effects on anti-Balb/c T cells in the MLR assays. In addition, the inoculation with Balb/c PerC B cells significantly prolonged the survival of subsequently grafted Balb/c hearts in B6 mouse recipients, whereas that with SPL B cells did not. These findings indicate that the PerC B cells, including PD-L1/PD-L2 B-1a cells, may suppress T cells responding to allostimulation, and thus may be optimal for donor lymphocyte injection.
Collapse
Affiliation(s)
- Takayuki Hirose
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Asuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Sakai
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yu Sasaki
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail:
| |
Collapse
|
24
|
Wang K, Tao L, Su J, Zhang Y, Zou B, Wang Y, Zou M, Chen N, Lei L, Li X. TLR4 supports the expansion of FasL +CD5 +CD1d hi regulatory B cells, which decreases in contact hypersensitivity. Mol Immunol 2017; 87:188-199. [PMID: 28505514 DOI: 10.1016/j.molimm.2017.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 04/18/2017] [Accepted: 04/23/2017] [Indexed: 02/06/2023]
Abstract
Certain B cells termed as "regulatory B cells" (Bregs) can suppress the ongoing immune responses and a splenic CD5+CD1dhi Breg subset identified earlier was shown to exert its regulatory functions through secretion of IL-10. Though FasL expression is an alternative mechanism of immune suppression used by B cells, little is known about the FasL expressing CD5+CD1dhi Bregs. In this study, we isolated splenocytes or splenic CD19+ B cells and compared the efficiency of toll-like receptor(TLR)4 ligand (lipopolysaccharide) with TLR9 ligand (CpG), anti-CD40 and TLR9 ligand (CpG) plus anti-CD40 on the FasL expression of splenic CD5+CD1dhi Bregs by flow cytometry. FasL expression in CD5+CD1dhi B cells was rapidly increased after TLR4 ligation. Intriguingly, anti-CD40 and CpG plus anti-CD40 combinations failed to stimulate FasL expression in CD5+CD1dhi B cells although the IL-10 production was up-regulated in this subset. In addition, LPS and other B10-cell inducers increased the expression of surface molecules like CD86 and CD25, which are correlated to the regulatory functions of B cells. Furthermore, NF-κB and NF-AT inhibitors decreased the TLR4-activated FasL expression in CD5+CD1dhi B cells. Then we sorted splenic CD5+CD1dhi Bregs using flow cytometry and found that TLR4-activated CD5+CD1dhi Bregs suppressed the proliferation of CFSE-labeled CD4+ T cells in vitro, which was partly blocked by anti-FasL antibody. In oxazolone-sensitized mice having contact hypersensitivity, FasL expression in splenic CD5+CD1dhi B cells was decreased compared to the control group after TLR4 ligation. Our findings suggest that the regulatory function of CD5+CD1dhi B cells could be partly mediated by Fas-FasL pathway and this FasL expressing CD5+CD1dhi Bregs might participate in the regulation of inflammatory diseases.
Collapse
Affiliation(s)
- Keng Wang
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Department of Clinical Pharmacy, The Affiliated Nanhai Hospital of Southern Medical University, Foshan 528200, PR China
| | - Lei Tao
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jianbing Su
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Yueyang Zhang
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Binhua Zou
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Yiyuan Wang
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Min Zou
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Nana Chen
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Linsheng Lei
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Xiaojuan Li
- Laboratory of Anti-inflammatory Immunomodulatory Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
25
|
van Rensburg IC, Kleynhans L, Keyser A, Walzl G, Loxton AG. B-cells with a FasL expressing regulatory phenotype are induced following successful anti-tuberculosis treatment. IMMUNITY INFLAMMATION AND DISEASE 2016; 5:57-67. [PMID: 28250925 PMCID: PMC5322165 DOI: 10.1002/iid3.140] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/20/2016] [Accepted: 10/25/2016] [Indexed: 12/19/2022]
Abstract
Introduction Studies show that B‐cells, in addition to producing antibodies and antigen‐presentation, are able to produce cytokines as well. These include regulatory cytokines such as IL‐10 by regulatory B‐cells. Furthermore, a rare regulatory subset of B‐cells have the potential to express FasL, which is a death‐inducing ligand. This subset of B‐cells have a positive role during autoimmune disease, but has not yet been studied during tuberculosis. These FasL‐expressing B‐cells are induced by bacterial LPS and CpG, thus we hypothesized that this phenotype might be induced during tuberculosis as well. Methods B‐cells from participants with TB (at diagnosis and during treatment) and controls were collected, and analyzed by means of real‐time PCR and flow cytometry. In addition to this, BAL was collected from TB participants as well and analyzed by means of MAGPix (multi‐cytokine) technology. Results Gene expression analysis show that FASL transcript levels increase by the end of treatment. Similarly, phenotypic analysis show that there is a higher frequency of FasL‐expressing B‐cells by the end of treatment. Conclusion Collectively, these results indicate that these FasL‐expressing B‐cells are being induced during anti‐TB treatment, and thus may play a positive role. Further studies are required to elucidate this.
Collapse
Affiliation(s)
- Ilana C van Rensburg
- Division of Molecular Biology and Human Genetics Faculty of Medicine and Health Sciences SA MRC Centre for TB Research DST/NRF Centre of Excellence for Biomedical Tuberculosis Research Stellenbosch University Cape Town South Africa
| | - Léanie Kleynhans
- Division of Molecular Biology and Human Genetics Faculty of Medicine and Health Sciences SA MRC Centre for TB Research DST/NRF Centre of Excellence for Biomedical Tuberculosis Research Stellenbosch University Cape Town South Africa
| | - Alana Keyser
- Clinical Laboratory Sciences Faculty of Health Sciences University of Cape Town Cape Town South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics Faculty of Medicine and Health Sciences SA MRC Centre for TB Research DST/NRF Centre of Excellence for Biomedical Tuberculosis Research Stellenbosch University Cape Town South Africa
| | - Andre G Loxton
- Division of Molecular Biology and Human Genetics Faculty of Medicine and Health Sciences SA MRC Centre for TB Research DST/NRF Centre of Excellence for Biomedical Tuberculosis Research Stellenbosch University Cape Town South Africa
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The number of deaths associated with cardiovascular disease remains high, despite great advances in treating the associated high levels of cholesterol. The main underlying pathology of cardiovascular disease is atherosclerosis, which is recognized as a chronic autoimmune-like inflammatory disease. Hence, there is a pressing need to shed light on the immune pathways associated with atherosclerosis. B cells have long been thought to have a general protective effect in atherosclerosis. However, findings in the last decade have challenged this paradigm, showing that it is crucial to differentiate between the various B-cell subsets when assessing their role/effect on atherosclerosis. RECENT FINDINGS It has become increasingly recognized lately that B cells can have significant effects on the immune system independent of antibody production. The understanding that B cells form a major source of cytokines and can directly influence T-cell responses via surface markers, have led to the identification of novel B-cell subsets. These subsets are important modulators of autoimmune disorders but have not yet been fully investigated in atherosclerosis. SUMMARY Here we review the current known roles of B-cell subsets and the putative effects of recently identified B cells on atherosclerosis.
Collapse
Affiliation(s)
- Hidde Douna
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
27
|
van de Veen W, Stanic B, Wirz OF, Jansen K, Globinska A, Akdis M. Role of regulatory B cells in immune tolerance to allergens and beyond. J Allergy Clin Immunol 2016; 138:654-665. [DOI: 10.1016/j.jaci.2016.07.006] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 12/21/2022]
|
28
|
Lundy SK, Wu Q, Wang Q, Dowling CA, Taitano SH, Mao G, Mao-Draayer Y. Dimethyl fumarate treatment of relapsing-remitting multiple sclerosis influences B-cell subsets. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e211. [PMID: 27006972 PMCID: PMC4784801 DOI: 10.1212/nxi.0000000000000211] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/22/2015] [Indexed: 12/04/2022]
Abstract
Objective: To test the hypothesis that dimethyl fumarate (Tecfidera, BG-12) affects B-cell subsets in patients with relapsing-remitting multiple sclerosis (RRMS). Methods: Peripheral blood B cells were compared for surface marker expression in patients with RRMS prior to initiation of treatment, after 4–6 months, and at more than 1 year of treatment with BG-12. Production of interleukin (IL)–10 by RRMS patient B cells was also analyzed. Results: Total numbers of peripheral blood B lymphocytes declined after 4–6 months of BG-12 treatment, due to losses in both the CD27+ memory B cells and CD27neg B-cell subsets. Some interpatient variability was observed. In contrast, circulating CD24highCD38high (T2-MZP) B cells increased in percentage in the majority of patients with RRMS after 4–6 months and were present in higher numbers in all of the patients after 12 months of treatment. The CD43+CD27+ B-1 B cells also increased at the later time point in most patients but were unchanged at 4–6 months compared to pretreatment levels. Purified B cells from 7 of the 9 patients with RRMS tested after 4–6 months of treatment were able to produce IL-10 following CD40 ligand stimulation, and the amount corresponded with the combined levels of T2-MZP and B-1 B cells in the sample. None of the patients with RRMS in this study have had a relapse while taking BG-12. Conclusions: These data suggest that BG-12 differentially affects B-cell subsets in patients with RRMS, resulting in increased numbers of circulating B lymphocytes with regulatory capacity.
Collapse
Affiliation(s)
- Steven K Lundy
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| | - Qi Wu
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| | - Qin Wang
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| | - Catherine A Dowling
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| | - Sophina H Taitano
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| | - Guangmei Mao
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| | - Yang Mao-Draayer
- Department of Internal Medicine, Division of Rheumatology (S.K.L.), Graduate Program in Immunology, Program in Biomedical Sciences (S.K.L., S.H.T., Y.M.-D.), and Department of Neurology (Q. Wu, Q. Wang, C.A.D., G.M., Y.M.-D.), University of Michigan Medical School, Ann Arbor
| |
Collapse
|
29
|
Tang Y, Jiang Q, Ou Y, Zhang F, Qing K, Sun Y, Lu W, Zhu H, Gong F, Lei P, Shen G. BIP induces mice CD19 hi regulatory B cells producing IL-10 and highly expressing PD-L1, FasL. Mol Immunol 2016; 69:44-51. [DOI: 10.1016/j.molimm.2015.10.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/13/2015] [Accepted: 10/26/2015] [Indexed: 12/15/2022]
|
30
|
Ray A, Wang L, Dittel BN. IL-10-independent regulatory B-cell subsets and mechanisms of action. Int Immunol 2015; 27:531-6. [PMID: 25999596 PMCID: PMC11513724 DOI: 10.1093/intimm/dxv033] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 05/18/2015] [Indexed: 12/16/2022] Open
Abstract
Although classically B cells are known to play important roles in immune protection via humoral immunity, recently their regulatory mechanisms have been best appreciated in the context of autoimmunity. Several studies have identified different subsets of regulatory B cells that vary not only in their phenotype but also in their mechanism of action. Although the best-studied mechanism of B-cell immune regulation is IL-10 production, other IL-10-independent mechanisms have been proposed. These include maintenance of CD4(+)Foxp3(+) regulatory T cells; production of transforming growth factor-β, IL-35, IgM or adenosine or expression of PD-L1 (programmed death 1 ligand 1) or FasL (Fas ligand). Given that B-cell-targeted therapy is being increasingly used in the clinic, a complete understanding of the mechanisms whereby B cells regulate inflammation associated with specific diseases is required for designing safe and effective immunotherapies targeting B cells.
Collapse
MESH Headings
- Animals
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/pathology
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Cell Lineage/immunology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fas Ligand Protein/genetics
- Fas Ligand Protein/immunology
- Gene Expression Regulation/immunology
- Graft vs Host Disease/genetics
- Graft vs Host Disease/immunology
- Graft vs Host Disease/pathology
- Humans
- Interleukin-10
- Interleukins/genetics
- Interleukins/immunology
- Mice
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/immunology
Collapse
Affiliation(s)
- Avijit Ray
- BloodCenter of Wisconsin, Blood Research Institute, Milwaukee, WI 53201, USA
| | - Luman Wang
- Department of Immunology and Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Biotherapy Research Center of Fudan University, Shanghai 200032, People's Republic of China
| | - Bonnie N Dittel
- BloodCenter of Wisconsin, Blood Research Institute, Milwaukee, WI 53201, USA
| |
Collapse
|
31
|
Kristensen B, Hegedüs L, Lundy SK, Brimnes MK, Smith TJ, Nielsen CH. Characterization of Regulatory B Cells in Graves' Disease and Hashimoto's Thyroiditis. PLoS One 2015; 10:e0127949. [PMID: 26016954 PMCID: PMC4446335 DOI: 10.1371/journal.pone.0127949] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 04/21/2015] [Indexed: 12/21/2022] Open
Abstract
A hallmark of regulatory B cells is IL-10 production, hence their designation as IL-10+ B cells. Little is known about the ability of self-antigens to induce IL-10+ B cells in Graves’ disease (GD), Hashimoto’s thyroiditis (HT), or other autoimmune disease. Here we pulsed purified B cells from 12 HT patients, 12 GD patients, and 12 healthy donors with the thyroid self-antigen, thyroglobulin (TG) and added the B cells back to the remaining peripheral blood mononuclear cells (PBMCs). This procedure induced IL-10+ B-cell differentiation in GD. A similar tendency was observed in healthy donors, but not in cells from patients with HT. In GD, B cells primed with TG induced IL-10-producing CD4+ T cells. To assess the maximal frequency of inducible IL-10+ B cells in the three donor groups PBMCs were stimulated with PMA/ionomycin. The resulting IL-10+ B-cell frequency was similar in the three groups and correlated with free T3 levels in GD patients. IL-10+ B cells from both patient groups displayed CD25 or TIM-1 more frequently than did those from healthy donors. B-cell expression of two surface marker combinations previously associated with regulatory B-cell functions, CD24hiCD38hi and CD27+CD43+, did not differ between patients and healthy donors. In conclusion, our findings indicate that autoimmune thyroiditis is not associated with reduced frequency of IL-10+ B cells. These results do not rule out regulatory B-cell dysfunction, however. The observed phenotypic differences between IL-10+ B cells from patients and healthy donors are discussed.
Collapse
Affiliation(s)
- Birte Kristensen
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
| | - Laszlo Hegedüs
- Department of Endocrinology and Metabolism, Odense University Hospital, Odense, Denmark
| | - Steven K. Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States of America
| | - Marie K. Brimnes
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Terry J. Smith
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center and Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Claus H. Nielsen
- Institute for Inflammation Research, Department of Infectious Diseases and Rheumatology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
32
|
Tao H, Lu L, Xia Y, Dai F, Wang Y, Bao Y, Lundy SK, Ito F, Pan Q, Zhang X, Zheng F, Shu G, Fang B, Jiang J, Xia J, Huang S, Li Q, Chang AE. Antitumor effector B cells directly kill tumor cells via the Fas/FasL pathway and are regulated by IL-10. Eur J Immunol 2015; 45:999-1009. [PMID: 25545618 PMCID: PMC4414939 DOI: 10.1002/eji.201444625] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 12/01/2014] [Accepted: 12/19/2014] [Indexed: 11/05/2022]
Abstract
We have previously reported that adoptive transfer of tumor-draining lymph node (TDLN) B cells confers tumor regression in a spontaneous pulmonary metastasis mouse model of breast cancer. In this study, we identified IL-10-producing cells within these B cells, and found that IL-10 removal, either by using IL-10(-/-) TDLN B cells or by systemic neutralization of IL-10, significantly augmented the therapeutic efficacy of adoptively transferred TDLN B cells. Depletion of IL-10 in B-cell adoptive transfers significantly increased CTLs and B-cell activity of PBMCs and splenic cells in the recipient. Activated TDLN B cells express Fas ligand, which was further enhanced by coculture of these TDLN B cells with 4T1 tumor cells. Effector B cells killed tumor cells directly in vitro in an antigen specific and Fas ligand-dependent manner. Trafficking of TDLN B cells in vivo suggested that they were recruited to the tumor and lung as well as secondary lymphoid organs. These findings further define the biological function of antitumor effector B cells, which may offer alternative cellular therapies to cancer.
Collapse
Affiliation(s)
- Huimin Tao
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yang Xia
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fu Dai
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Wang
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyi Bao
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Steven K. Lundy
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fumito Ito
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Qin Pan
- Wuhan University School of Medicine, Wuhan, China
| | | | - Fang Zheng
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoshun Shu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- Second Xiangya Hospital, Changsha, China
| | - Bingmu Fang
- Lishui People’s Hospital of Zhejiang Province, China
| | - Jinhong Jiang
- Lishui People’s Hospital of Zhejiang Province, China
| | - Jianchuang Xia
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shiang Huang
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Alfred E. Chang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
33
|
Lundy SK, Klinker MW, Fox DA. Killer B lymphocytes and their fas ligand positive exosomes as inducers of immune tolerance. Front Immunol 2015; 6:122. [PMID: 25852690 PMCID: PMC4367442 DOI: 10.3389/fimmu.2015.00122] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/04/2015] [Indexed: 01/31/2023] Open
Abstract
Induction of immune tolerance is a key process by which the immune system is educated to modulate reactions against benign stimuli such as self-antigens and commensal microbes. Understanding and harnessing the natural mechanisms of immune tolerance may become an increasingly useful strategy for treating many types of allergic and autoimmune diseases, as well as for improving the acceptance of solid organ transplants. Our laboratory and others have been interested in the natural ability of some B lymphocytes to express the death-inducing molecule Fas ligand (FasL), and their ability to kill T helper (TH) lymphocytes. We have recently shown that experimental transformation of human B cells by a non-replicative variant of Epstein-Barr virus (EBV) consistently resulted in high expression of functional FasL protein. The production and release of FasL+ exosomes that co-expressed major histocompatibility complex (MHC) class II molecules and had the capacity to kill antigen-specific TH cells was also observed. Several lines of evidence indicate that FasL+ B cells and FasL+MHCII+ exosomes have important roles in natural immune tolerance and have a great deal of therapeutic potential. Taken together, these findings suggest that EBV-immortalized human B lymphoblastoid cell lines could be used as cellular factories for FasL+ exosomes, which would be employed to therapeutically establish and/or regain immune tolerance toward specific antigens. The goals of this review are to summarize current knowledge of the roles of FasL+ B cells and exosomes in immune regulation, and to suggest methods of manipulating killer B cells and FasL+ exosomes for clinical purposes.
Collapse
Affiliation(s)
- Steven K Lundy
- Department of Internal Medicine-Rheumatology, University of Michigan Medical School , Ann Arbor, MI , USA ; Graduate Training Program in Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - Matthew W Klinker
- Graduate Training Program in Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| | - David A Fox
- Department of Internal Medicine-Rheumatology, University of Michigan Medical School , Ann Arbor, MI , USA ; Graduate Training Program in Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
34
|
Lundy SK, Klinker MW. Characterization and activity of Fas ligand producing CD5⁺ B cells. Methods Mol Biol 2015; 1190:81-102. [PMID: 25015275 DOI: 10.1007/978-1-4939-1161-5_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
B lymphocytes make several contributions to immune regulation including production of antibodies with regulatory properties, release of immune suppressive cytokines, and expression of death-inducing ligands. A role for Fas ligand (FasL)-expressing "killer" B cells in regulating T helper cell survival and chronic inflammation has been demonstrated in animal models of schistosome worm infection, asthma, and autoimmune arthritis. Interestingly, a population of CD5(+) B cells found in the spleen and lungs of naïve mice constitutively expresses FasL and has potent killer function against T helper cells that is antigen-specific and FasL-dependent. Killer B cells therefore represent a novel target for immune modulation in many disease settings. Our laboratory has recently published methods of characterizing FasL(+) B cells and inducing their proliferation in vitro. This chapter will describe detailed methods of identifying and expanding killer B cells from mice, detecting FasL expression in B cells, and performing functional killing assays against antigen-specific TH cells.
Collapse
Affiliation(s)
- Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, 4043 Biomedical Sciences Research Bldg., 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA,
| | | |
Collapse
|
35
|
Role of dendritic cells in the initiation, progress and modulation of systemic autoimmune diseases. Autoimmun Rev 2015; 14:127-39. [DOI: 10.1016/j.autrev.2014.10.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 09/30/2014] [Indexed: 12/11/2022]
|
36
|
Braza F, Chesne J, Castagnet S, Magnan A, Brouard S. Regulatory functions of B cells in allergic diseases. Allergy 2014; 69:1454-63. [PMID: 25060230 DOI: 10.1111/all.12490] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 12/24/2022]
Abstract
B cells are essentially described for their capacity to produce antibodies ensuring anti-infectious immunity or deleterious responses in the case of autoimmunity or allergy. However, abundant data described their ability to restrain inflammation by diverse mechanisms. In allergy, some regulatory B-cell subsets producing IL-10 have been recently described as potent suppressive cells able to restrain inflammatory responses both in vitro and in vivo by regulatory T-cell differentiation or directly inhibiting T-cell-mediated inflammation. A specific deficit in regulatory B cells participates to more severe allergic inflammation. Induction of allergen tolerance through specific immunotherapy induces a specific expansion of these cells supporting their role in establishment of allergen tolerance. However, the regulatory functions carried out by B cells are not exclusively IL-10 dependent. Indeed, other regulatory mechanisms mediated by B cells are (i) the production of TGF-β, (ii) the promotion of T-cell apoptosis by Fas-Fas ligand or granzyme-B pathways, and (iii) their capacity to produce inhibitory IgG4 and sialylated IgG able to mediate anti-inflammatory mechanisms. This points to Bregs as interesting targets for the development of new therapies to induce allergen tolerance. In this review, we highlight advances in the study of regulatory mechanisms mediated by B cells and outline what is known about their phenotype as well as their suppressive role in allergy from studies in both mice and humans.
Collapse
Affiliation(s)
- F. Braza
- INSERM; UMR 1087; l'institut du Thorax; Nantes France
- CNRS; UMR 6291; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- INSERM; UMR U1064; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
| | - J. Chesne
- INSERM; UMR 1087; l'institut du Thorax; Nantes France
- CNRS; UMR 6291; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- INSERM; UMR U1064; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
| | - S. Castagnet
- Laboratoire HLA; Établissement Français du Sang; Nantes France
| | - A. Magnan
- INSERM; UMR 1087; l'institut du Thorax; Nantes France
- CNRS; UMR 6291; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
- CHU Nantes; l'institut du Thorax; Service de Pneumologie; Nantes France
| | - S. Brouard
- INSERM; UMR U1064; Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu; Nantes France
- Université de Nantes; Nantes France
| |
Collapse
|
37
|
Klinker MW, Lizzio V, Reed TJ, Fox DA, Lundy SK. Human B Cell-Derived Lymphoblastoid Cell Lines Constitutively Produce Fas Ligand and Secrete MHCII(+)FasL(+) Killer Exosomes. Front Immunol 2014; 5:144. [PMID: 24765093 PMCID: PMC3980107 DOI: 10.3389/fimmu.2014.00144] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 03/20/2014] [Indexed: 12/18/2022] Open
Abstract
Immune suppression mediated by exosomes is an emerging concept with potentially immense utility for immunotherapy in a variety of inflammatory contexts, including allogeneic transplantation. Exosomes containing the apoptosis-inducing molecule Fas ligand (FasL) have demonstrated efficacy in inhibiting antigen-specific immune responses upon adoptive transfer in animal models. We report here that a very high frequency of human B cell-derived lymphoblastoid cell lines (LCL) constitutively produce MHCII+FasL+ exosomes that can induce apoptosis in CD4+ T cells. All LCL tested for this study (>20 independent cell lines) showed robust expression of FasL, but had no detectable FasL on the cell surface. Given this intracellular sequestration, we hypothesized that FasL in LCL was retained in the secretory lysosome and secreted via exosomes. Indeed, we found both MHCII and FasL proteins present in LCL-derived exosomes, and using a bead-based exosome capture assay demonstrated the presence of MHCII+FasL+ exosomes among those secreted by LCL. Using two independent experimental approaches, we demonstrated that LCL-derived exosomes were capable of inducing antigen-specific apoptosis in autologous CD4+ T cells. These results suggest that LCL-derived exosomes may present a realistic source of immunosuppressive exosomes that could reduce or eliminate T cell-mediated responses against donor-derived antigens in transplant recipients.
Collapse
Affiliation(s)
- Matthew W Klinker
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Vincent Lizzio
- Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Tamra J Reed
- Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - David A Fox
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| | - Steven K Lundy
- Graduate Program in Immunology, University of Michigan , Ann Arbor, MI , USA ; Division of Rheumatology, Department of Internal Medicine, University of Michigan , Ann Arbor, MI , USA
| |
Collapse
|
38
|
|