1
|
Cho SH, Jones MA, Meyer K, Anderson DM, Chetyrkin S, Calcutt MW, Caprioli RM, Semenkovich CF, Boothby MR. B cell expression of the enzyme PexRAP, an intermediary in ether lipid biosynthesis, promotes antibody responses and germinal center size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618760. [PMID: 39464149 PMCID: PMC11507954 DOI: 10.1101/2024.10.17.618760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The qualities of antibody (Ab) responses provided by B lymphocytes and their plasma cell (PC) descendants are crucial facets of responses to vaccines and microbes. Metabolic processes and products regulate aspects of B cell proliferation and differentiation into germinal center (GC) and PC states as well as Ab diversification. However, there is little information about lymphoid cell-intrinsic functions of enzymes that mediate ether lipid biosynthesis, including a major class of membrane phospholipids. Imaging mass spectrometry (IMS) results had indicated that concentrations of a number of these phospholipids were substantially enhanced in GC compared to the background average in spleens. However, it was not clear if biosynthesis in B cells was a basis for this finding, or whether such cell-intrinsic biosynthesis contributes to B cell physiology or Ab responses. Ether lipid biosynthesis can involve the enzyme PexRAP, the product of the Dhrs7b gene. Using combinations of IMS and immunization experiments in mouse models with inducible Dhrs7b loss-of-function, we now show that B lineage-intrinsic expression of PexRAP promotes the magnitude and affinity maturation of a serological response. Moreover, the data revealed a Dhrs7b-dependent increase in ether phospholipids in primary follicles with a more prominent increase in GC. Mechanistically, PexRAP impacted B cell proliferation via enhanced survival associated with controlling levels of ROS and membrane peroxidation. These findings reveal a vital role of this peroxisomal enzyme in B cell homeostasis and the physiology of humoral immunity.
Collapse
|
2
|
Ding Q, Wu Y, Triglia ET, Gommerman JL, Subramanian A, Kuchroo VK, Rothstein DM. TIM-4 Identifies Effector B Cells Expressing a RORγt-Driven Proinflammatory Cytokine Module That Promotes Immune Responsiveness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.22.558524. [PMID: 37790513 PMCID: PMC10542535 DOI: 10.1101/2023.09.22.558524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
B cells can express pro-inflammatory cytokines that promote a wide variety of immune responses. Here we show that B cells expressing the phosphatidylserine receptor TIM-4, preferentially express IL-17A, as well as IL-22, IL-6, IL-1β, and GM-CSF - a collection of cytokines reminiscent of pathogenic Th17 cells. Expression of this proinflammatory module requires IL-23R signaling and selective expression of RORγt and IL-17A by TIM-4+ B cells. TIM-4+ B cell-derived-IL-17A not only enhances the severity of experimental autoimmune encephalomyelitis (EAE) and promotes allograft rejection, but also acts in an autocrine manner to prevent their conversion into IL-10-expressing B cells with regulatory function. Thus, IL-17A acts as an inflammatory mediator and also enforces the proinflammatory activity of TIM-4+ B cells. Thus, TIM-4 serves as a broad marker for RORγt+ effector B cells (Beff) and allows further study of the signals regulating Beff differentiation and effector molecule expression.
Collapse
Affiliation(s)
- Qing Ding
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yufan Wu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Elena Torlai Triglia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | | | - Ayshwarya Subramanian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- The Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Vijay K. Kuchroo
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA, USA
- The Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - David M. Rothstein
- Thomas E. Starzl Transplantation Institute; University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| |
Collapse
|
3
|
Jiang H, Wang S. Physical extraction of antigen and information. Proc Natl Acad Sci U S A 2024; 121:e2320537121. [PMID: 39302963 PMCID: PMC11441497 DOI: 10.1073/pnas.2320537121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 07/18/2024] [Indexed: 09/22/2024] Open
Abstract
To respond and adapt, cells use surface receptors to sense environmental cues. While biochemical signal processing inside the cell is studied in depth, less is known about how physical processes during cell-cell contact impact signal acquisition. New experiments found that fast-evolving immune B cells in germinal centers (GCs) apply force to acquire antigen clusters prior to internalization, suggesting adaptive benefits of physical information extraction. We present a theory of stochastic antigen transfer and show that maximizing information gain via physical extraction can explain the dramatic phenotypic transition from naive to GC B cells-attenuated receptor signaling, enhanced force usage, and decentralized contact architecture. Our model suggests that binding-lifetime measurement and physical extraction serve as complementary modes of antigen recognition, greatly extending the dynamic range of affinity discrimination when combined. This physical-information framework further predicts that the optimal size of receptor clusters decreases as affinity improves, rationalizing the use of a multifocal synaptic pattern seen in GC B cells. By linking extraction dynamics to selection fidelity via discriminatory performance, we propose that cells may physically enhance information acquisition to sustain adaptive evolution.
Collapse
Affiliation(s)
- Hongda Jiang
- Department of Physics and Astronomy, University of California, Los Angeles, CA90095
| | - Shenshen Wang
- Department of Physics and Astronomy, University of California, Los Angeles, CA90095
| |
Collapse
|
4
|
Degn SE, Tolar P. Towards a unifying model for B-cell receptor triggering. Nat Rev Immunol 2024:10.1038/s41577-024-01073-x. [PMID: 39256626 DOI: 10.1038/s41577-024-01073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 09/12/2024]
Abstract
Antibodies are exceptionally versatile molecules with remarkable flexibility in their binding properties. Their natural targets range from small-molecule toxins, across viruses of different sizes, to bacteria and large multicellular parasites. The molecular determinants bound by antibodies include proteins, peptides, carbohydrates, nucleic acids, lipids and even synthetic molecules that have never existed in nature. Membrane-anchored antibodies also serve as receptors on the surface of the B cells that produce them. Despite recent structural insights, there is still no unifying molecular mechanism to explain how antibody targets (antigens) trigger the activation of these B-cell receptors (BCRs). After cognate antigen encounter, somatic hypermutation and class-switch recombination allow BCR affinity maturation and immunoglobulin class-specific responses, respectively. This raises the fundamental question of how one receptor activation mechanism can accommodate a plethora of variant receptors and ligands, and how it can ensure that individual B cells remain responsive to antigen after somatic hypermutation and class switching. There is still no definite answer. Here we give a brief historical account of the different models proposed to explain BCR triggering and discuss their merit in the context of the current knowledge of the structure of BCRs, their dynamic membrane distribution, and recent biochemical and cell biological insights.
Collapse
Affiliation(s)
- Søren E Degn
- Laboratory for Lymphocyte Biology, Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Centre for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark.
| | - Pavel Tolar
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
5
|
Inoue T, Baba Y, Kurosaki T. BCR signaling in germinal center B cell selection. Trends Immunol 2024; 45:693-704. [PMID: 39168721 DOI: 10.1016/j.it.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024]
Abstract
When mature B cells are activated by antigens, the selection of these activated B cells takes place particularly during T cell-dependent immune responses in which an improved antibody repertoire is generated through somatic hypermutation in germinal centers (GCs). In this process the importance of antigen presentation by GC B cells, and subsequent T follicular helper (Tfh) cell help in positive selection of GC B cells, has been well appreciated. By contrast, the role of B cell receptor (BCR) signaling per se remains unclear. Strong experimental support for the involvement of BCR signaling in GC B cell selection has now been provided. Interestingly, these studies suggest that several checkpoints operating through the BCR ensure affinity maturation.
Collapse
Affiliation(s)
- Takeshi Inoue
- Department of Molecular Systems Immunology, University of Tokyo Pandemic Preparedness, Infection, and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, Osaka, Japan; Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan; Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan.
| |
Collapse
|
6
|
Hong Y, Kwak K. Both sides now: evolutionary traits of antigens and B cells in tolerance and activation. Front Immunol 2024; 15:1456220. [PMID: 39185403 PMCID: PMC11341355 DOI: 10.3389/fimmu.2024.1456220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 08/27/2024] Open
Abstract
B cells are the cornerstone of our body's defense system, producing precise antibodies and safeguarding immunological memory for future protection against pathogens. While we have a thorough understanding of how naïve B cells differentiate into plasma or memory B cells, the early B cell response to various antigens-whether self or foreign-remains a thrilling and evolving area of study. Advances in imaging have illuminated the molecular intricacies of B cell receptor (BCR) signaling, yet the dynamic nature of B cell activation continues to reveal new insights based on the nature of antigen exposure. This review explores the evolutionary journey of B cells as they adapt to the unique challenges presented by pathogens. We begin by examining the specific traits of antigens that influence their pathogenic potential, then shift our focus to the distinct characteristics of B cells that counteract these threats. From foundational discoveries to the latest cutting-edge research, we investigate how B cells are effectively activated and distinguish between self and non-self antigens, ensuring a balanced immune response that defends against pathogenic diseases but not self-antigens.
Collapse
Affiliation(s)
- Youngjae Hong
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Kihyuck Kwak
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Huang H, Narayanan HV, Hoffmann A. Synergy and antagonism in the integration of BCR and CD40 signals that control B-cell proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.28.605521. [PMID: 39131345 PMCID: PMC11312454 DOI: 10.1101/2024.07.28.605521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
In response to infection or vaccination, a successful antibody response must enrich high-affinity antigen-reactive B-cells through positive selection, but eliminate auto-reactive B-cells through negative selection. B-cells receive signals from the B-cell receptor (BCR) which binds the antigen, and the CD40 receptor which is stimulated by neighboring T-cells that also recognize the antigen. How BCR and CD40 signaling are integrated quantitatively to jointly determine B-cell fate decision and proliferation remains unclear. To investigate this, we developed a differential-equations-based model of the BCR and CD40 signaling networks activating NFκB. Our model accurately recapitulates the NFκB dynamics of B-cells stimulated through their BCR and CD40 receptors, correctly predicting that costimulation induces more NFκB activity. However, when linking it to established cell fate decision models of cell survival and cell cycle control, it predicted potentiated population expansion that was not observed experimentally. We found that this discrepancy was due to a time-dependent functional antagonism exacerbated by BCR-induced caspase activity that can trigger apoptosis in founder cells, unless NFκB-induced survival gene expression protects B-cells in time. Guided by model predictions, sequential co-stimulation experiments revealed how the temporal dynamics of BCR and CD40 signaling control the fate decision between negative and positive selection of B-cell clonal expansion. Our quantitative findings highlight a complex non-monotonic integration of BCR and CD40 signals that is controlled by a balance between NFκB and cell-death pathways, and suggest a mechanism for regulating the stringency of B-cell selection during an antibody response.
Collapse
Affiliation(s)
- Helen Huang
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics (MIMG)
- Institute for Quantitative and Computational Biosciences (QCBio)
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, USA
| | - Haripriya Vaidehi Narayanan
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics (MIMG)
- Institute for Quantitative and Computational Biosciences (QCBio)
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics (MIMG)
- Institute for Quantitative and Computational Biosciences (QCBio)
| |
Collapse
|
8
|
Schiepers A, Van't Wout MFL, Hobbs A, Mesin L, Victora GD. Opposing effects of pre-existing antibody and memory T cell help on the dynamics of recall germinal centers. Immunity 2024; 57:1618-1628.e4. [PMID: 38838672 PMCID: PMC11236515 DOI: 10.1016/j.immuni.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/20/2024] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
Re-exposure to an antigen generates abundant antibody responses and drives the formation of secondary germinal centers (GCs). Recall GCs in mice consist almost entirely of naïve B cells, whereas recall antibodies derive overwhelmingly from memory B cells. Here, we examine this division between cellular and serum compartments. After repeated immunization with the same antigen, tetramer analyses of recall GCs revealed a marked decrease in the ability of B cells in these structures to bind the antigen. Boosting with viral variant proteins restored antigen binding in recall GCs, as did genetic ablation of primary-derived antibody-secreting cells through conditional deletion of Prdm1, demonstrating suppression of GC recall responses by pre-existing antibodies. In hapten-carrier experiments in which B and T cell specificities were uncoupled, memory T cell help allowed B cells with undetectable antigen binding to access GCs. Thus, antibody-mediated feedback steers recall GC B cells away from previously targeted epitopes and enables specific targeting of variant epitopes, with implications for vaccination protocols.
Collapse
Affiliation(s)
- Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | | | - Alvaro Hobbs
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
9
|
Inoue T, Matsumoto Y, Kawai C, Ito M, Nada S, Okada M, Kurosaki T. Csk restrains BCR-mediated ROS production and contributes to germinal center selection and affinity maturation. J Exp Med 2024; 221:e20231996. [PMID: 38753246 PMCID: PMC11098938 DOI: 10.1084/jem.20231996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/26/2024] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
Compared with naïve B cells, the B cell receptor (BCR) signal in germinal center (GC) B cells is attenuated; however, the significance of this signaling attenuation has not been well defined. Here, to investigate the role of attenuation of BCR signaling, we employed a Csk mutant mouse model in which Csk deficiency in GC B cells resulted in augmentation of net BCR signaling with no apparent effect on antigen presentation. We found that Csk is required for GC maintenance and efficient antibody affinity maturation. Mechanistically, ROS-induced apoptosis was exacerbated concomitantly with mitochondrial dysfunction in Csk-deficient GC B cells. Hence, our data suggest that attenuation of the BCR signal restrains hyper-ROS production, thereby protecting GC B cells from apoptosis and contributing to efficient affinity maturation.
Collapse
Affiliation(s)
- Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Molecular Systems Immunology, The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Yuma Matsumoto
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Chie Kawai
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Mao Ito
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shigeyuki Nada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masato Okada
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| |
Collapse
|
10
|
Fiske BE, Wemlinger SM, Crute BW, Getahun A. The Src-family kinase Lyn plays a critical role in establishing and maintaining B cell anergy by suppressing PI3K-dependent signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595208. [PMID: 38826354 PMCID: PMC11142063 DOI: 10.1101/2024.05.21.595208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Although the Src family kinase (SFK) Lyn is known to be involved in induction and maintenance of peripheral B cell tolerance, the molecular basis of its action in this context remains unclear. This question has been approached using conventional as well as B cell-targeted knockouts of Lyn, with varied conclusions likely confused by collateral loss of Lyn functions in B cell and myeloid cell development and activation. Here we utilized a system in which Lyn gene deletion is tamoxifen inducible and B cell restricted. This system allows acute elimination of Lyn in B cells without off-target effects. This genetic tool was employed in conjunction with immunoglobulin transgenic mice in which peripheral B cells are autoreactive. DNA reactive Ars/A1 B cells require continuous inhibitory signaling, mediated by the inositol phosphatase SHIP-1 and the tyrosine phosphatase SHP-1, to maintain an unresponsive (anergic) state. Here we show that Ars/A1 B cells require Lyn to establish and maintain B cell unresponsiveness. Lyn primarily functions by restricting PI3K-dependent signaling pathways. This Lyn-dependent mechanism complements the impact of reduced mIgM BCR expression to restrict BCR signaling in Ars/A1 B cells. Our findings suggest that a subset of autoreactive B cells requires Lyn to become anergic and that the autoimmunity associated with dysregulated Lyn function may, in part, be due to an inability of these autoreactive B cells to become tolerized.
Collapse
|
11
|
Zhang L, Toboso-Navasa A, Gunawan A, Camara A, Nakagawa R, Katja F, Chakravarty P, Newman R, Zhang Y, Eilers M, Wack A, Tolar P, Toellner KM, Calado DP. Regulation of BCR-mediated Ca 2+ mobilization by MIZ1-TMBIM4 safeguards IgG1 + GC B cell-positive selection. Sci Immunol 2024; 9:eadk0092. [PMID: 38579014 PMCID: PMC7615907 DOI: 10.1126/sciimmunol.adk0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/26/2024] [Indexed: 04/07/2024]
Abstract
The transition from immunoglobulin M (IgM) to affinity-matured IgG antibodies is vital for effective humoral immunity. This is facilitated by germinal centers (GCs) through affinity maturation and preferential maintenance of IgG+ B cells over IgM+ B cells. However, it is not known whether the positive selection of the different Ig isotypes within GCs is dependent on specific transcriptional mechanisms. Here, we explored IgG1+ GC B cell transcription factor dependency using a CRISPR-Cas9 screen and conditional mouse genetics. We found that MIZ1 was specifically required for IgG1+ GC B cell survival during positive selection, whereas IgM+ GC B cells were largely independent. Mechanistically, MIZ1 induced TMBIM4, an ancestral anti-apoptotic protein that regulated inositol trisphosphate receptor (IP3R)-mediated calcium (Ca2+) mobilization downstream of B cell receptor (BCR) signaling in IgG1+ B cells. The MIZ1-TMBIM4 axis prevented mitochondrial dysfunction-induced IgG1+ GC cell death caused by excessive Ca2+ accumulation. This study uncovers a unique Ig isotype-specific dependency on a hitherto unidentified mechanism in GC-positive selection.
Collapse
Affiliation(s)
- Lingling Zhang
- Immunity and Cancer, Francis Crick Institute, London, UK
| | | | - Arief Gunawan
- Immunity and Cancer, Francis Crick Institute, London, UK
| | | | | | | | | | - Rebecca Newman
- Immune Receptor Activation Laboratory, Francis Crick Institute, London, UK
| | - Yang Zhang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Martin Eilers
- Theodor Boveri Institute and Comprehensive Cancer Center Mainfranken, Biocenter, University of Würzburg, Würzburg, Germany
| | | | - Pavel Tolar
- Immune Receptor Activation Laboratory, Francis Crick Institute, London, UK
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | |
Collapse
|
12
|
Fiske BE, Getahun A. Failed Downregulation of PI3K Signaling Makes Autoreactive B Cells Receptive to Bystander T Cell Help. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1150-1160. [PMID: 38353615 PMCID: PMC10948302 DOI: 10.4049/jimmunol.2300108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 01/16/2024] [Indexed: 02/27/2024]
Abstract
The role of T cell help in autoantibody responses is not well understood. Because tolerance mechanisms govern both T and B cell responses, one might predict that both T cell tolerance and B cell tolerance must be defeated in autoantibody responses requiring T cell help. To define whether autoreactive B cells depend on T cells to generate autoantibody responses, we studied the role of T cells in murine autoantibody responses resulting from acute B cell-specific deletion of regulatory phosphatases. Ars/A1 B cells are DNA reactive and require continuous inhibitory signaling by the tyrosine phosphatase SHP-1 and the inositol phosphatases SHIP-1 and PTEN to maintain unresponsiveness. Acute B cell-restricted deletion of any of these phosphatases results in an autoantibody response. In this study, we show that CD40-CD40L interactions are required to support autoantibody responses of B cells whose anergy has been compromised. If the B cell-intrinsic driver of loss of tolerance is failed negative regulation of PI3K signaling, bystander T cells provide sufficient CD40-mediated signal 2 to support an autoantibody response. However, although autoantibody responses driven by acute B cell-targeted deletion of SHP-1 also require T cells, bystander T cell help does not suffice. These results demonstrate that upregulation of PI3K signaling in autoreactive B cells, recapitulating the effect of multiple autoimmunity risk alleles, promotes autoantibody responses both by increasing B cells' cooperation with noncognate T cell help and by altering BCR signaling. Receptiveness to bystander T cell help enables autoreactive B cells to circumvent the fail-safe of T cell tolerance.
Collapse
Affiliation(s)
- Brigita E. Fiske
- Department of Immunology and Microbiology, University of Colorado SOM, Aurora, CO, USA
| | - Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado SOM, Aurora, CO, USA
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| |
Collapse
|
13
|
Sutton HJ, Gao X, Kelly HG, Parker BJ, Lofgren M, Dacon C, Chatterjee D, Seder RA, Tan J, Idris AH, Neeman T, Cockburn IA. Lack of affinity signature for germinal center cells that have initiated plasma cell differentiation. Immunity 2024; 57:245-255.e5. [PMID: 38228150 PMCID: PMC10922795 DOI: 10.1016/j.immuni.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/08/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024]
Abstract
Long-lived plasma cells (PCs) secrete antibodies that can provide sustained immunity against infection. High-affinity cells are proposed to preferentially select into this compartment, potentiating the immune response. We used single-cell RNA-seq to track the germinal center (GC) development of Ighg2A10 B cells, specific for the Plasmodium falciparum circumsporozoite protein (PfCSP). Following immunization with Plasmodium sporozoites, we identified 3 populations of cells in the GC light zone (LZ). One LZ population expressed a gene signature associated with the initiation of PC differentiation and readily formed PCs in vitro. The estimated affinity of these pre-PC B cells was indistinguishable from that of LZ cells that remained in the GC. This remained true when high- or low-avidity recombinant PfCSP proteins were used as immunogens. These findings suggest that the initiation of PC development occurs via an affinity-independent process.
Collapse
Affiliation(s)
- Henry J Sutton
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Xin Gao
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Hannah G Kelly
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Brian J Parker
- Biological Data Science Institute, The Australian National University, Canberra, ACT 2601, Australia; School of Computing, ANU College of Engineering, Computing & Cybernetics, The Australian National University, Canberra, ACT 2601, Australia
| | - Mariah Lofgren
- Malaria Unit, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cherrelle Dacon
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Deepyan Chatterjee
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
| | - Robert A Seder
- Malaria Unit, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joshua Tan
- Antibody Biology Unit, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Azza H Idris
- Malaria Unit, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Department of Pediatrics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Teresa Neeman
- Biological Data Science Institute, The Australian National University, Canberra, ACT 2601, Australia
| | - Ian A Cockburn
- Division of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia.
| |
Collapse
|
14
|
Ishikawa M, Hasanali ZS, Zhao Y, Das A, Lavaert M, Roman CJ, Londregan J, Allman D, Bhandoola A. Bone marrow plasma cells require P2RX4 to sense extracellular ATP. Nature 2024; 626:1102-1107. [PMID: 38355795 PMCID: PMC11025016 DOI: 10.1038/s41586-024-07047-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 01/09/2024] [Indexed: 02/16/2024]
Abstract
Plasma cells produce large quantities of antibodies and so play essential roles in immune protection1. Plasma cells, including a long-lived subset, reside in the bone marrow where they depend on poorly defined microenvironment-linked survival signals1. We show that bone marrow plasma cells use the ligand-gated purinergic ion channel P2RX4 to sense extracellular ATP released by bone marrow osteoblasts through the gap-junction protein pannexin 3 (PANX3). Mutation of Panx3 or P2rx4 each caused decreased serum antibodies and selective loss of bone marrow plasma cells. Compared to their wild-type counterparts, PANX3-null osteoblasts secreted less extracellular ATP and failed to support plasma cells in vitro. The P2RX4-specific inhibitor 5-BDBD abrogated the impact of extracellular ATP on bone marrow plasma cells in vitro, depleted bone marrow plasma cells in vivo and reduced pre-induced antigen-specific serum antibody titre with little posttreatment rebound. P2RX4 blockade also reduced autoantibody titre and kidney disease in two mouse models of humoral autoimmunity. P2RX4 promotes plasma cell survival by regulating endoplasmic reticulum homeostasis, as short-term P2RX4 blockade caused accumulation of endoplasmic reticulum stress-associated regulatory proteins including ATF4 and B-lineage mutation of the pro-apoptotic ATF4 target Chop prevented bone marrow plasma cell demise on P2RX4 inhibition. Thus, generating mature protective and pathogenic plasma cells requires P2RX4 signalling controlled by PANX3-regulated extracellular ATP release from bone marrow niche cells.
Collapse
Affiliation(s)
- Masaki Ishikawa
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4254, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104-6082, USA
| | - Zainul S. Hasanali
- Address correspondence to: Masaki Ishikawa () David Allman (), or Avinash Bhandoola ()
| | - Yongge Zhao
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4254, USA
| | - Arundhoti Das
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4254, USA
| | - Marieke Lavaert
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4254, USA
| | | | | | - David Allman
- Address correspondence to: Masaki Ishikawa () David Allman (), or Avinash Bhandoola ()
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-4254, USA
| |
Collapse
|
15
|
Zhou JZ, Huang B, Pei B, Sun GW, Pawlitz MD, Zhang W, Li X, Hokynar KC, Yao F, Perera MLW, Wei S, Zheng S, Polin LA, Poulik JM, Ranki A, Krohn K, Cunningham-Rundles C, Yang N, Bhagwat AS, Yu K, Peterson P, Kisand K, Vuong BQ, Cerutti A, Chen K. A Germinal Center Checkpoint of AIRE in B Cells Limits Antibody Diversification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.574926. [PMID: 38260362 PMCID: PMC10802573 DOI: 10.1101/2024.01.10.574926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
In response to antigens, B cells undergo affinity maturation and class switching mediated by activation-induced cytidine deaminase (AID) in germinal centers (GCs) of secondary lymphoid organs, but uncontrolled AID activity can precipitate autoimmunity and cancer. The regulation of GC antibody diversification is of fundamental importance but not well understood. We found that autoimmune regulator (AIRE), the molecule essential for T cell tolerance, is expressed in GC B cells in a CD40-dependent manner, interacts with AID and negatively regulates antibody affinity maturation and class switching by inhibiting AID function. AIRE deficiency in B cells caused altered antibody repertoire, increased somatic hypermutations, elevated autoantibodies to T helper 17 effector cytokines and defective control of skin Candida albicans. These results define a GC B cell checkpoint of humoral immunity and illuminate new approaches of generating high-affinity neutralizing antibodies for immunotherapy.
Collapse
Affiliation(s)
- Jordan Z. Zhou
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- These authors contributed equally
| | - Bihui Huang
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
- These authors contributed equally
| | - Bo Pei
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Guang Wen Sun
- School of Applied Science, Republic Polytechnic, Singapore 738984, Singapore
| | - Michael D. Pawlitz
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Wei Zhang
- Beijing Genomics Institute (BGI)-Shenzhen, Guangdong 518083, China
| | - Xinyang Li
- Beijing Genomics Institute (BGI)-Shenzhen, Guangdong 518083, China
| | - Kati C. Hokynar
- Department of Virology, University of Helsinki, Helsinki 00029, Finland
| | - Fayi Yao
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | | | - Shanqiao Wei
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA
| | - Simin Zheng
- School of Biological Sciences, Nanyang Technological University, Singapore 636921, Singapore
| | - Lisa A. Polin
- Barbara Ann Karmanos Cancer Institute, Department of Oncology, Wayne State University, Detroit, MI 48201, USA
| | - Janet M. Poulik
- Department of Pathology, Children’s Hospital of Michigan, Detroit, MI 48201, USA
| | - Annamari Ranki
- Department of Dermatology and Allergic Diseases, University of Helsinki and Helsinki University Hospital, Helsinki 00250, Finland
| | - Kai Krohn
- Helsinki University Hospital Research Institute, Biomedicum, Helsinki 00290, Finland
| | | | - Naibo Yang
- Beijing Genomics Institute (BGI)-Shenzhen, Guangdong 518083, China
- Complete Genomics Inc., Mountain View, California 94043, USA
| | - Ashok S. Bhagwat
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University, Detroit, MI 48201, USA
| | - Kefei Yu
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Pärt Peterson
- Department of Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Kai Kisand
- Department of Molecular Pathology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu 50411, Estonia
| | - Bao Q. Vuong
- Department of Biology, City College of New York, New York, NY 10031, USA
| | - Andrea Cerutti
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mucosal Immunology Studies Team, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Maryland 20892, USA
| | - Kang Chen
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- School of Biological Sciences, Nanyang Technological University, Singapore 636921, Singapore
- Lead Contact
| |
Collapse
|
16
|
Dominguez-Sola D. Sending positive signals and good (calcium) vibes. J Exp Med 2024; 221:e20231821. [PMID: 38051276 PMCID: PMC10697794 DOI: 10.1084/jem.20231821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
In this issue of JEM, Yada et al. (https://doi.org/10.1084/jem.20222178) demonstrate that effective antibody affinity selection in germinal centers relies on the store-operated calcium entry (SOCE) component of the B cell receptor (BCR) signaling network. Therefore, active BCR signaling is as relevant to positive selection as the function of BCRs as endocytic receptors, answering a question that had puzzled experts for a while. These findings transform our understanding of the mechanisms supporting adaptive immune responses (to vaccines, for example) and have important implications for interpreting the genomics and pathogenesis of germinal center-derived B cell lymphomas.
Collapse
Affiliation(s)
- David Dominguez-Sola
- Departments of Oncological Sciences and Pathology, The Tisch Cancer Institute, Marc and Jennifer Lipschultz Precision Immunology Institute, Center for Advanced Blood Cancer Therapies and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
17
|
Brookens SK, Cho SH, Paik Y, Meyer K, Raybuck AL, Park C, Greenwood DL, Rathmell JC, Boothby MR. Plasma Cell Differentiation, Antibody Quality, and Initial Germinal Center B Cell Population Depend on Glucose Influx Rate. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:43-56. [PMID: 37955416 PMCID: PMC10841396 DOI: 10.4049/jimmunol.2200756] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/19/2023] [Indexed: 11/14/2023]
Abstract
Serum Ab concentrations, selection for higher affinity BCRs, and generation of higher Ab affinities are important elements of immune response optimization and functions of germinal center (GC) reactions. B cell proliferation requires nutrients to support the anabolism inherent in clonal expansion. Glucose usage by mouse GC B cells has been reported to contribute little to their energy needs, with questions raised as to whether glucose uptake or glycolysis increases in GC B cells compared with their naive precursors. Indeed, metabolism can be highly flexible, such that supply shortage along one pathway may be compensated by increased flux on others. We now show that reduction of the glucose transporter GLUT1 in mice after establishment of a preimmune B cell repertoire, even after initiation of the GC B cell gene expression program, decreased initial GC B cell population numbers, affinity maturation, and plasma cell outputs. Glucose oxidation was heightened in GC B cells, but this hexose flowed more into the pentose phosphate pathway, whose activity was important in controlling reactive oxygen species (ROS) and Ab-secreting cell production. In modeling how glucose usage by B cells promotes the Ab response, the control of ROS appeared insufficient. Surprisingly, the combination of galactose, which mitigated ROS, with provision of mannose, an efficient precursor to glycosylation, supported robust production of and normal Ab secretion by Ab-secreting cells under glucose-free conditions. Collectively, the findings indicate that GCs depend on normal glucose influx, especially in plasma cell production, but reveal an unexpected metabolic flexibility in hexose requirements.
Collapse
Affiliation(s)
- Shawna K. Brookens
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Cancer Biology Program, Vanderbilt University
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA 19104
| | - Sung Hoon Cho
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Inflammation, & Immunology
| | - Yeeun Paik
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Kaylor Meyer
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Ariel L. Raybuck
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Chloe Park
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Dalton L. Greenwood
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Jeffrey C. Rathmell
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Cancer Biology Program, Vanderbilt University
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Inflammation, & Immunology
| | - Mark R. Boothby
- Department of Pathology-Microbiology-Immunology, Vanderbilt University Medical Center, Nashville, TN 37232
- Cancer Biology Program, Vanderbilt University
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Inflammation, & Immunology
| |
Collapse
|
18
|
Yada Y, Matsumoto M, Inoue T, Baba A, Higuchi R, Kawai C, Yanagisawa M, Kitamura D, Ohga S, Kurosaki T, Baba Y. STIM-mediated calcium influx regulates maintenance and selection of germinal center B cells. J Exp Med 2024; 221:e20222178. [PMID: 37902601 PMCID: PMC10615893 DOI: 10.1084/jem.20222178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/02/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023] Open
Abstract
Positive selection of high-affinity germinal center (GC) B cells is driven by antigen internalization through their B cell receptor (BCR) and presentation to follicular helper T cells. However, the requirements of BCR signaling in GC B cells remain poorly understood. Store-operated Ca2+ entry, mediated by stromal interacting molecule 1 (STIM1) and STIM2, is the main Ca2+ influx pathway triggered by BCR engagement. Here, we showed that STIM-deficient B cells have reduced B cell competitiveness compared with wild-type B cells during GC responses. B cell-specific deletion of STIM proteins decreased the number of high-affinity B cells in the late phase of GC formation. STIM deficiency did not affect GC B cell proliferation and antigen presentation but led to the enhancement of apoptosis due to the impaired upregulation of anti-apoptotic Bcl2a1. STIM-mediated activation of NFAT was required for the expression of Bcl2a1 after BCR stimulation. These findings suggest that STIM-mediated survival signals after antigen capture regulate the optimal selection and maintenance of GC B cells.
Collapse
Affiliation(s)
- Yutaro Yada
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masanori Matsumoto
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akemi Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryota Higuchi
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Chie Kawai
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Daisuke Kitamura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoshihiro Baba
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
19
|
Schiepers A, van ‘t Wout MFL, Hobbs A, Mesin L, Victora GD. Opposing effects of pre-existing antibody and memory T cell help on the dynamics of recall germinal centers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.571936. [PMID: 38168231 PMCID: PMC10760098 DOI: 10.1101/2023.12.15.571936] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Re-exposure to an antigen generates serum antibody responses that greatly exceed in magnitude those elicited by primary antigen encounter, while simultaneously driving the formation of recall germinal centers (GCs). Although recall GCs in mice are composed almost entirely of naïve B cells, recall antibody titers derive overwhelmingly from memory B cells, suggesting a division between cellular and serum compartments. Here, we show that this schism is at least partly explained by a marked decrease in the ability of recall GC B cells to detectably bind antigen. Variant priming and plasmablast ablation experiments show that this decrease is largely due to suppression by pre-existing antibody, whereas hapten-carrier experiments reveal a role for memory T cell help in allowing B cells with undetectable antigen binding to access GCs. We propose a model in which antibody-mediated feedback steers recall GC B cells away from previously targeted epitopes, thus enabling specific targeting of variant epitopes.
Collapse
Affiliation(s)
- Ariën Schiepers
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | | | - Alvaro Hobbs
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Gabriel D. Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
20
|
Roy K, Chakraborty M, Kumar A, Manna AK, Roy NS. The NFκB signaling system in the generation of B-cell subsets: from germinal center B cells to memory B cells and plasma cells. Front Immunol 2023; 14:1185597. [PMID: 38169968 PMCID: PMC10758606 DOI: 10.3389/fimmu.2023.1185597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024] Open
Abstract
Memory B cells and antibody-secreting cells are the two prime effector B cell populations that drive infection- and vaccine-induced long-term antibody-mediated immunity. The antibody-mediated immunity mostly relies on the formation of specialized structures within secondary lymphoid organs, called germinal centers (GCs), that facilitate the interactions between B cells, T cells, and antigen-presenting cells. Antigen-activated B cells may proliferate and differentiate into GC-independent plasmablasts and memory B cells or differentiate into GC B cells. The GC B cells undergo proliferation coupled to somatic hypermutation of their immunoglobulin genes for antibody affinity maturation. Subsequently, affinity mature GC B cells differentiate into GC-dependent plasma cells and memory B cells. Here, we review how the NFκB signaling system controls B cell proliferation and the generation of GC B cells, plasmablasts/plasma cells, and memory B cells. We also identify and discuss some important unanswered questions in this connection.
Collapse
Affiliation(s)
- Koushik Roy
- Division of Microbiology and Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Mainak Chakraborty
- Division of Immunology, Indian Council of Medical Research-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Ashok Kumar
- Division of Microbiology and Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Asit Kumar Manna
- Division of Microbiology and Immunology, Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, United States
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Neeladri Sekhar Roy
- Department of Biochemistry, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
21
|
Sprumont A, Rodrigues A, McGowan SJ, Bannard C, Bannard O. Germinal centers output clonally diverse plasma cell populations expressing high- and low-affinity antibodies. Cell 2023; 186:5486-5499.e13. [PMID: 37951212 DOI: 10.1016/j.cell.2023.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/05/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Germinal centers (GCs) form in lymph nodes after immunization or infection to facilitate antibody affinity maturation and memory and plasma cell (PC) development. PC differentiation is thought to involve stringent selection for GC B cells expressing the highest-affinity antigen receptors, but how this plays out during complex polyclonal responses is unclear. We combine temporal lineage tracing with antibody characterization to gain a snapshot of PCs developing during influenza infection. GCs co-mature B cell clones with antibody affinities spanning multiple orders of magnitude; however, each generates PCs with similar efficiencies, including weak binders. Within lineages, PC selection is not restricted to variants with the highest-affinity antibodies. Differentiation is commonly associated with proliferative expansion to produce "nodes" of identical PCs. Immunization-induced GCs generate fewer PCs but still of low- and high-antibody affinities. We propose that generating low-affinity antibody PCs reflects an evolutionary compromise to facilitate diverse serum antibody responses.
Collapse
Affiliation(s)
- Adrien Sprumont
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Ana Rodrigues
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Simon J McGowan
- Computational Biology Research Group, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Colin Bannard
- Department of Linguistics and English Language, University of Manchester, Manchester M13 9PL, UK
| | - Oliver Bannard
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.
| |
Collapse
|
22
|
Tilstra JS, Kim M, Gordon RA, Leibler C, Cosgrove HA, Bastacky S, Nickerson KM, Shlomchik MJ. B cell-intrinsic Myd88 regulates disease progression in murine lupus. J Exp Med 2023; 220:e20230263. [PMID: 37787782 PMCID: PMC10541815 DOI: 10.1084/jem.20230263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/26/2023] [Accepted: 09/11/2023] [Indexed: 10/04/2023] Open
Abstract
Nucleic acid-specific Toll-like receptors (TLRs) have been implicated in promoting disease pathogenesis in systemic lupus erythematosus (SLE). Whether such TLRs mediate disease onset, progression, or both remains undefined; yet the answer to this question has important therapeutic implications. MyD88 is an essential adaptor that acts downstream of IL-1 family receptors and most TLRs. Both global and B cell-specific Myd88 deficiency ameliorated disease in lupus-prone mice when constitutively deleted. To address whether Myd88 was needed to sustain ongoing disease, we induced B cell-specific deletion of Myd88 after disease onset in MRL.Faslpr mice using an inducible Cre recombinase. B cell-specific deletion of Myd88 starting after disease onset resulted in ameliorated glomerulonephritis and interstitial inflammation. Additionally, treated mice had reduced autoantibody formation and an altered B cell compartment with reduced ABC and plasmablast numbers. These experiments demonstrate the role of MyD88 in B cells to sustain disease in murine lupus. Therefore, targeting MyD88 or its upstream activators may be a viable therapeutic option in SLE.
Collapse
Affiliation(s)
- Jeremy S. Tilstra
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Lupus Center of Excellence, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Minjung Kim
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rachael A. Gordon
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Claire Leibler
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haylee A. Cosgrove
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sheldon Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin M. Nickerson
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mark J. Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Bauer-Smith H, Sudol ASL, Beers SA, Crispin M. Serum immunoglobulin and the threshold of Fc receptor-mediated immune activation. Biochim Biophys Acta Gen Subj 2023; 1867:130448. [PMID: 37652365 PMCID: PMC11032748 DOI: 10.1016/j.bbagen.2023.130448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 09/02/2023]
Abstract
Antibodies can mediate immune recruitment or clearance of immune complexes through the interaction of their Fc domain with cellular Fc receptors. Clustering of antibodies is a key step in generating sufficient avidity for efficacious receptor recognition. However, Fc receptors may be saturated with prevailing, endogenous serum immunoglobulin and this raises the threshold by which cellular receptors can be productively engaged. Here, we review the factors controlling serum IgG levels in both healthy and disease states, and discuss how the presence of endogenous IgG is encoded into the functional activation thresholds for low- and high-affinity Fc receptors. We discuss the circumstances where antibody engineering can help overcome these physiological limitations of therapeutic antibodies. Finally, we discuss how the pharmacological control of Fc receptor saturation by endogenous IgG is emerging as a feasible mechanism for the enhancement of antibody therapeutics.
Collapse
Affiliation(s)
- Hannah Bauer-Smith
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK
| | - Abigail S L Sudol
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Stephen A Beers
- Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton SO16 6YD, UK.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| |
Collapse
|
24
|
Wang PH, Washburn RS, Mariuzza DL, Lin WHW, Gill AL, Ahmed R, Reiner SL. Reciprocal transmission of activating and inhibitory signals and cell fate in regenerating T cells. Cell Rep 2023; 42:113155. [PMID: 37756164 PMCID: PMC10872930 DOI: 10.1016/j.celrep.2023.113155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/14/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
The ability of activated progenitor T cells to self-renew while producing differentiated effector cell descendants may underlie immunological memory and persistent responses to ongoing infection. The nature of stem-like T cells responding to cancer and during treatment with immunotherapy is not clear. The subcellular organization of dividing progenitor CD8+ T cells from mice challenged with syngeneic tumors is examined here. Three-dimensional microscopy reveals an activating hub composed of polarized CD3, CD28, and phosphatidylinositol 3-kinase (PI3K) activity at the putative immunological synapse with an inhibitory hub composed of polarized PD-1 and CD73 at the opposite pole of mitotic blasts. Progenitor T cells from untreated and inhibitory checkpoint blockade-treated mice yield a differentiated TCF1- daughter cell, which inherits the PI3K activation hub, alongside a discordantly fated, self-renewing TCF1+ sister cell. Dynamic organization of opposite activating and inhibitory signaling poles in mitotic lymphocytes may account for the enigmatic durability of specific immunity.
Collapse
Affiliation(s)
- Peter H Wang
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Robert S Washburn
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dylan L Mariuzza
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Wen-Hsuan W Lin
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Amanda L Gill
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Steven L Reiner
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
25
|
Raso F, Liu S, Simpson MJ, Barton GM, Mayer CT, Acharya M, Muppidi JR, Marshak-Rothstein A, Reboldi A. Antigen receptor signaling and cell death resistance controls intestinal humoral response zonation. Immunity 2023; 56:2373-2387.e8. [PMID: 37714151 PMCID: PMC10591993 DOI: 10.1016/j.immuni.2023.08.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/24/2023] [Accepted: 08/21/2023] [Indexed: 09/17/2023]
Abstract
Immunoglobulin A (IgA) maintains commensal communities in the intestine while preventing dysbiosis. IgA generated against intestinal microbes assures the simultaneous binding to multiple, diverse commensal-derived antigens. However, the exact mechanisms by which B cells mount broadly reactive IgA to the gut microbiome remains elusive. Here, we have shown that IgA B cell receptor (BCR) is required for B cell fitness during the germinal center (GC) reaction in Peyer's patches (PPs) and for generation of gut-homing plasma cells (PCs). We demonstrate that IgA BCR drove heightened intracellular signaling in mouse and human B cells, and as a consequence, IgA+ B cells received stronger positive selection cues. Mechanistically, IgA BCR signaling offset Fas-mediated death, possibly rescuing low-affinity B cells to promote a broad humoral response to commensals. Our findings reveal an additional mechanism linking BCR signaling, B cell fate, and antibody production location, which have implications for how intestinal antigen recognition shapes humoral immunity.
Collapse
Affiliation(s)
- Fiona Raso
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shuozhi Liu
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Mikala J Simpson
- Experimental Immunology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Gregory M Barton
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Christian T Mayer
- Experimental Immunology Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Mridu Acharya
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Jagan R Muppidi
- Lymphoid Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Ann Marshak-Rothstein
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
26
|
Brookens SK, Cho SH, Paik Y, Meyer K, Raybuck AL, Park C, Greenwood DL, Rathmell JC, Boothby MR. Plasma cell differentiation, antibody quality, and initial germinal center B cell population depend on glucose influx rate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.13.557599. [PMID: 37745429 PMCID: PMC10515901 DOI: 10.1101/2023.09.13.557599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Antibody secretion into sera, selection for higher affinity BCR, and the generation of higher Ab affinities are important elements of immune response optimization, and a core function of germinal center reactions. B cell proliferation requires nutrients to support the anabolism inherent in clonal expansion. Glucose usage by GC B cells has been reported to contribute little to their energy needs, with questions raised as to whether or not glucose uptake or glycolysis increases in GC B cells compared to their naïve precursors. Indeed, metabolism can be highly flexible, such that supply shortage along one pathway may be compensated by increased flux on others. We now show that elimination of the glucose transporter GLUT1 after establishment of a pre-immune B cell repertoire, even after initiation of the GC B cell gene expression program, decreased initial GC B cell population numbers, affinity maturation, and PC outputs. Glucose oxidation was heightened in GC B cells, but this hexose flowed more into the pentose phosphate pathway (PPP), whose activity was important in controlling reactive oxygen (ROS) and ASC production. In modeling how glucose usage by B cells promotes the Ab response, the control of ROS appeared insufficient. Surprisingly, the combination of galactose, which mitigated ROS, with provision of mannose - an efficient precursor to glycosylation - supported robust production of and normal Ab secretion by ASC under glucose-free conditions. Collectively, the findings indicate that GC depend on normal glucose influx, especially in PC production, but reveal an unexpected metabolic flexibility in hexose requirements. KEY POINTS Glucose influx is critical for GC homeostasis, affinity maturation and the generation of Ab-secreting cells.Plasma cell development uses the Pentose Phosphate Pathway, and hexose sugars maintain redox homeostasis.PCs can develop and achieve robust Ab secretion in the absence of glucose using a combination of hexose alternatives.
Collapse
|
27
|
Guldenpfennig C, Teixeiro E, Daniels M. NF-kB's contribution to B cell fate decisions. Front Immunol 2023; 14:1214095. [PMID: 37533858 PMCID: PMC10391175 DOI: 10.3389/fimmu.2023.1214095] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/03/2023] [Indexed: 08/04/2023] Open
Abstract
NF-κB signaling is essential to an effective innate and adaptive immune response. Many immune-specific functional and developmental outcomes depend in large on NF-κB. The formidable task of sorting out the mechanisms behind the regulation and outcome of NF-κB signaling remains an important area of immunology research. Here we briefly discuss the role of NF-κB in regulating cell fate decisions at various times in the path of B cell development, activation, and the generation of long-term humoral immunity.
Collapse
Affiliation(s)
- Caitlyn Guldenpfennig
- Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Emma Teixeiro
- Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| | - Mark Daniels
- Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
| |
Collapse
|
28
|
Bod L, Kye YC, Shi J, Torlai Triglia E, Schnell A, Fessler J, Ostrowski SM, Von-Franque MY, Kuchroo JR, Barilla RM, Zaghouani S, Christian E, Delorey TM, Mohib K, Xiao S, Slingerland N, Giuliano CJ, Ashenberg O, Li Z, Rothstein DM, Fisher DE, Rozenblatt-Rosen O, Sharpe AH, Quintana FJ, Apetoh L, Regev A, Kuchroo VK. B-cell-specific checkpoint molecules that regulate anti-tumour immunity. Nature 2023; 619:348-356. [PMID: 37344597 PMCID: PMC10795478 DOI: 10.1038/s41586-023-06231-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/17/2023] [Indexed: 06/23/2023]
Abstract
The role of B cells in anti-tumour immunity is still debated and, accordingly, immunotherapies have focused on targeting T and natural killer cells to inhibit tumour growth1,2. Here, using high-throughput flow cytometry as well as bulk and single-cell RNA-sequencing and B-cell-receptor-sequencing analysis of B cells temporally during B16F10 melanoma growth, we identified a subset of B cells that expands specifically in the draining lymph node over time in tumour-bearing mice. The expanding B cell subset expresses the cell surface molecule T cell immunoglobulin and mucin domain 1 (TIM-1, encoded by Havcr1) and a unique transcriptional signature, including multiple co-inhibitory molecules such as PD-1, TIM-3, TIGIT and LAG-3. Although conditional deletion of these co-inhibitory molecules on B cells had little or no effect on tumour burden, selective deletion of Havcr1 in B cells both substantially inhibited tumour growth and enhanced effector T cell responses. Loss of TIM-1 enhanced the type 1 interferon response in B cells, which augmented B cell activation and increased antigen presentation and co-stimulation, resulting in increased expansion of tumour-specific effector T cells. Our results demonstrate that manipulation of TIM-1-expressing B cells enables engagement of the second arm of adaptive immunity to promote anti-tumour immunity and inhibit tumour growth.
Collapse
Affiliation(s)
- Lloyd Bod
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yoon-Chul Kye
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jingwen Shi
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- BeiGene, Beijing, China
| | - Elena Torlai Triglia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandra Schnell
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Johannes Fessler
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | | | - Max Y Von-Franque
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| | - Juhi R Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Rocky M Barilla
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sarah Zaghouani
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Elena Christian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Toni Marie Delorey
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kanishka Mohib
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sheng Xiao
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Nadine Slingerland
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David M Rothstein
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Department of Biology and Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arlene H Sharpe
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Francisco J Quintana
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Department of Biology and Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lionel Apetoh
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- INSERM, Tours, France
- Faculté de Médecine, Université de Tours, Tours, France
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Department of Biology and Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Genentech, San Francisco, CA, USA.
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
O'Connor RS, May MJ, Freedman BD. Opening Orai's to see B-yond current paradigms. Cell Calcium 2023; 113:102758. [PMID: 37209447 PMCID: PMC10804139 DOI: 10.1016/j.ceca.2023.102758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/22/2023]
Affiliation(s)
- Roderick S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd., Philadelphia, PA 19104
| | - Michael J May
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania
| | - Bruce D Freedman
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104.
| |
Collapse
|
30
|
Chandiran K, Cauley LS. The diverse effects of transforming growth factor-β and SMAD signaling pathways during the CTL response. Front Immunol 2023; 14:1199671. [PMID: 37426662 PMCID: PMC10327426 DOI: 10.3389/fimmu.2023.1199671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play an important role in defense against infections with intracellular pathogens and anti-tumor immunity. Efficient migration is required to locate and destroy infected cells in different regions of the body. CTLs accomplish this task by differentiating into specialized subsets of effector and memory CD8 T cells that traffic to different tissues. Transforming growth factor-beta (TGFβ) belongs to a large family of growth factors that elicit diverse cellular responses via canonical and non-canonical signaling pathways. Canonical SMAD-dependent signaling pathways are required to coordinate changes in homing receptor expression as CTLs traffic between different tissues. In this review, we discuss the various ways that TGFβ and SMAD-dependent signaling pathways shape the cellular immune response and transcriptional programming of newly activated CTLs. As protective immunity requires access to the circulation, emphasis is placed on cellular processes that are required for cell-migration through the vasculature.
Collapse
Affiliation(s)
- Karthik Chandiran
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Linda S. Cauley
- Department of Immunology, UCONN Health, Farmington, CT, United States
| |
Collapse
|
31
|
Pasqualucci L. The germinal center in the pathogenesis of B cell lymphomas. Hematol Oncol 2023; 41 Suppl 1:62-69. [PMID: 37294970 DOI: 10.1002/hon.3141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
The adaptive immune system has evolved to allow effective responses against a virtually unlimited number of invading pathogens. This process requires the transient formation of germinal centers (GC), a dynamic environment that ensures the generation and selection of B cells capable to produce antibodies with high antigen affinity, or to maintain the memory of that antigen for life. However, this comes at a cost, as the unique events accompanying the GC reaction pose a significant risk to the genome of B cells, which must endure elevated levels of replication stress, while proliferating at high rates and undergoing DNA breaks introduced by somatic hypermutation and class switch recombination. Indeed, the genetic/epigenetic disruption of programs implicated in normal GC biology has emerged as a hallmark of most B cell lymphomas. This improved understanding provides a conceptual framework for the identification of cellular pathways that could be exploited for precision medicine approaches.
Collapse
Affiliation(s)
- Laura Pasqualucci
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, and the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, USA
| |
Collapse
|
32
|
Fike AJ, Chodisetti SB, Wright NE, Bricker KN, Domeier PP, Maienschein-Cline M, Rosenfeld AM, Luckenbill SA, Weber JL, Choi NM, Luning Prak ET, Mandal M, Clark MR, Rahman ZSM. STAT3 signaling in B cells controls germinal center zone organization and recycling. Cell Rep 2023; 42:112512. [PMID: 37200190 PMCID: PMC10311431 DOI: 10.1016/j.celrep.2023.112512] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/05/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023] Open
Abstract
Germinal centers (GCs), sites of antibody affinity maturation, are organized into dark (DZ) and light (LZ) zones. Here, we show a B cell-intrinsic role for signal transducer and activator of transcription 3 (STAT3) in GC DZ and LZ organization. Altered zonal organization of STAT3-deficient GCs dampens development of long-lived plasma cells (LL-PCs) but increases memory B cells (MBCs). In an abundant antigenic environment, achieved here by prime-boost immunization, STAT3 is not required for GC initiation, maintenance, or proliferation but is important for sustaining GC zonal organization by regulating GC B cell recycling. Th cell-derived signals drive STAT3 tyrosine 705 and serine 727 phosphorylation in LZ B cells, regulating their recycling into the DZ. RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analyses identified STAT3 regulated genes that are critical for LZ cell recycling and transiting through DZ proliferation and differentiation phases. Thus, STAT3 signaling in B cells controls GC zone organization and recycling, and GC egress of PCs, but negatively regulates MBC output.
Collapse
Affiliation(s)
- Adam J Fike
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Sathi Babu Chodisetti
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nathaniel E Wright
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL 60637, USA
| | - Kristen N Bricker
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Phillip P Domeier
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | | | - Aaron M Rosenfeld
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara A Luckenbill
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Julia L Weber
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Nicholas M Choi
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Eline T Luning Prak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malay Mandal
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL 60637, USA
| | - Marcus R Clark
- Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL 60637, USA
| | - Ziaur S M Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
33
|
Chen ST, Oliveira TY, Gazumyan A, Cipolla M, Nussenzweig MC. B cell receptor signaling in germinal centers prolongs survival and primes B cells for selection. Immunity 2023; 56:547-561.e7. [PMID: 36882061 PMCID: PMC10424567 DOI: 10.1016/j.immuni.2023.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 10/28/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023]
Abstract
Germinal centers (GCs) are sites of B cell clonal expansion, diversification, and antibody affinity selection. This process is limited and directed by T follicular helper cells that provide helper signals to B cells that endocytose, process, and present cognate antigens in proportion to their B cell receptor (BCR) affinity. Under this model, the BCR functions as an endocytic receptor for antigen capture. How signaling through the BCR contributes to selection is not well understood. To investigate the role of BCR signaling in GC selection, we developed a tracker for antigen binding and presentation and a Bruton's tyrosine kinase drug-resistant-mutant mouse model. We showed that BCR signaling per se is necessary for the survival and priming of light zone B cells to receive T cell help. Our findings provide insight into how high-affinity antibodies are selected within GCs and are fundamental to our understanding of adaptive immunity and vaccine development.
Collapse
Affiliation(s)
- Spencer T Chen
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA.
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Melissa Cipolla
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute (HHMI), The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
34
|
Luo W, Conter L, Elsner RA, Smita S, Weisel F, Callahan D, Wu S, Chikina M, Shlomchik M. IL-21R signal reprogramming cooperates with CD40 and BCR signals to select and differentiate germinal center B cells. Sci Immunol 2023; 8:eadd1823. [PMID: 36800413 PMCID: PMC10206726 DOI: 10.1126/sciimmunol.add1823] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023]
Abstract
Both B cell receptor (BCR) and CD40 signaling are rewired in germinal center (GC) B cells (GCBCs) to synergistically induce c-MYC and phosphorylated S6 ribosomal protein (p-S6), markers of positive selection. How interleukin-21 (IL-21), a key T follicular helper (TFH)-derived cytokine, affects GCBCs is unclear. Like BCR and CD40 signals, IL-21 receptor (IL-21R) plus CD40 signals also synergize to induce c-MYC and p-S6 in GCBCs. However, IL-21R plus CD40 stimulation differentially affects GCBC fate compared with BCR plus CD40 ligation-engaging unique molecular mechanisms-as revealed by bulk RNA sequencing (RNA-seq), single-cell RNA-seq, and flow cytometry of GCBCs in vitro and in vivo. Whereas both signal pairs induced BLIMP1 in some GCBCs, only the IL-21R/CD40 combination induced IRF4hi/CD138+ cells, indicative of plasma cell differentiation, along with CCR6+/CD38+ memory B cell precursors. These findings reveal a second positive selection pathway in GCBCs, document rewired IL-21R signaling in GCBCs, and link specific TFH- and Ag-derived signals to GCBC differentiation.
Collapse
Affiliation(s)
- Wei Luo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- These authors contributed equally
- Present address: Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Laura Conter
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- These authors contributed equally
| | - Rebecca A. Elsner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- These authors contributed equally
| | - Shuchi Smita
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Florian Weisel
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Derrick Callahan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Shuxian Wu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Maria Chikina
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Mark Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Lead contact
| |
Collapse
|
35
|
Chen Z, Cui Y, Yao Y, Liu B, Yunis J, Gao X, Wang N, Cañete PF, Tuong ZK, Sun H, Wang H, Yang S, Wang R, Leong YA, Simon Davis D, Qin J, Liang K, Deng J, Wang CK, Huang YH, Roco JA, Nettelfield S, Zhu H, Xu H, Yu Z, Craik D, Liu Z, Qi H, Parish C, Yu D. Heparan sulfate regulates IL-21 bioavailability and signal strength that control germinal center B cell selection and differentiation. Sci Immunol 2023; 8:eadd1728. [PMID: 36800411 DOI: 10.1126/sciimmunol.add1728] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
In antibody responses, mutated germinal center B (BGC) cells are positively selected for reentry or differentiation. As the products from GCs, memory B cells and antibody-secreting cells (ASCs) support high-affinity and long-lasting immunity. Positive selection of BGC cells is controlled by signals received through the B cell receptor (BCR) and follicular helper T (TFH) cell-derived signals, in particular costimulation through CD40. Here, we demonstrate that the TFH cell effector cytokine interleukin-21 (IL-21) joins BCR and CD40 in supporting BGC selection and reveal that strong IL-21 signaling prioritizes ASC differentiation in vivo. BGC cells, compared with non-BGC cells, show significantly reduced IL-21 binding and attenuated signaling, which is mediated by low cellular heparan sulfate (HS) sulfation. Mechanistically, N-deacetylase and N-sulfotransferase 1 (Ndst1)-mediated N-sulfation of HS in B cells promotes IL-21 binding and signal strength. Ndst1 is down-regulated in BGC cells and up-regulated in ASC precursors, suggesting selective desensitization to IL-21 in BGC cells. Thus, specialized biochemical regulation of IL-21 bioavailability and signal strength sets a balance between the stringency and efficiency of GC selection.
Collapse
Affiliation(s)
- Zhian Chen
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Yanfang Cui
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, Central China Normal University, Wuhan, China
| | - Yin Yao
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Department of Otolaryngology-Head and Neck Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing, China
| | - Joseph Yunis
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Xin Gao
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Naiqi Wang
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Pablo F Cañete
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK.,Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Hongjian Sun
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Hao Wang
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Siling Yang
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Runli Wang
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Yew Ann Leong
- Centre for Inflammatory Diseases, Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Melbourne, VIC, Australia
| | - David Simon Davis
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jiahuan Qin
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaili Liang
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Deng
- China-Australia Centre for Personalised Immunology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Conan K Wang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD, Australia
| | - Yen-Hua Huang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Jonathan A Roco
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Sam Nettelfield
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Huaming Zhu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Huajun Xu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Zhijia Yu
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - David Craik
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, University of Queensland, Brisbane, QLD, Australia
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, Tsinghua University, Beijing, China
| | - Christopher Parish
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Di Yu
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia.,Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Emrich SM, Yoast RE, Zhang X, Fike AJ, Wang YH, Bricker KN, Tao AY, Xin P, Walter V, Johnson MT, Pathak T, Straub AC, Feske S, Rahman ZSM, Trebak M. Orai3 and Orai1 mediate CRAC channel function and metabolic reprogramming in B cells. eLife 2023; 12:e84708. [PMID: 36803766 PMCID: PMC9998091 DOI: 10.7554/elife.84708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
The essential role of store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels in T cells is well established. In contrast, the contribution of individual Orai isoforms to SOCE and their downstream signaling functions in B cells are poorly understood. Here, we demonstrate changes in the expression of Orai isoforms in response to B cell activation. We show that both Orai3 and Orai1 mediate native CRAC channels in B cells. The combined loss of Orai1 and Orai3, but not Orai3 alone, impairs SOCE, proliferation and survival, nuclear factor of activated T cells (NFAT) activation, mitochondrial respiration, glycolysis, and the metabolic reprogramming of primary B cells in response to antigenic stimulation. Nevertheless, the combined deletion of Orai1 and Orai3 in B cells did not compromise humoral immunity to influenza A virus infection in mice, suggesting that other in vivo co-stimulatory signals can overcome the requirement of BCR-mediated CRAC channel function in B cells. Our results shed important new light on the physiological roles of Orai1 and Orai3 proteins in SOCE and the effector functions of B lymphocytes.
Collapse
Affiliation(s)
- Scott M Emrich
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Ryan E Yoast
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Xuexin Zhang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Adam J Fike
- Department of Microbiology and Immunology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Yin-Hu Wang
- Department of Pathology, New York University School of MedicineNew YorkUnited States
| | - Kristen N Bricker
- Department of Microbiology and Immunology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Anthony Y Tao
- Department of Pathology, New York University School of MedicineNew YorkUnited States
| | - Ping Xin
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburghUnited States
- Vascular Medicine Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Vonn Walter
- Department of Public Health Sciences, Pennsylvania State University College of MedicineHersheyUnited States
| | - Martin T Johnson
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Trayambak Pathak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburghUnited States
- Vascular Medicine Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Adam C Straub
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburghUnited States
- Vascular Medicine Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Stefan Feske
- Department of Pathology, New York University School of MedicineNew YorkUnited States
| | - Ziaur SM Rahman
- Department of Microbiology and Immunology, Pennsylvania State University College of MedicineHersheyUnited States
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of MedicineHersheyUnited States
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of MedicinePittsburghUnited States
- Vascular Medicine Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| |
Collapse
|
37
|
Targeting protein phosphatases in cancer immunotherapy and autoimmune disorders. Nat Rev Drug Discov 2023; 22:273-294. [PMID: 36693907 PMCID: PMC9872771 DOI: 10.1038/s41573-022-00618-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 01/25/2023]
Abstract
Protein phosphatases act as key regulators of multiple important cellular processes and are attractive therapeutic targets for various diseases. Although extensive effort has been dedicated to phosphatase-targeted drug discovery, early expeditions for competitive phosphatase inhibitors were plagued by druggability issues, leading to the stigmatization of phosphatases as difficult targets. Despite challenges, persistent efforts have led to the identification of several drug-like, non-competitive modulators of some of these enzymes - including SH2 domain-containing protein tyrosine phosphatase 2, protein tyrosine phosphatase 1B, vascular endothelial protein tyrosine phosphatase and protein phosphatase 1 - reigniting interest in therapeutic targeting of phosphatases. Here, we discuss recent progress in phosphatase drug discovery, with emphasis on the development of selective modulators that exhibit biological activity. The roles and regulation of protein phosphatases in immune cells and their potential as powerful targets for immuno-oncology and autoimmunity indications are assessed.
Collapse
|
38
|
Fiske BE, Getahun A. Failed down-regulation of PI3K signaling makes autoreactive B cells receptive to bystander T cell help. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525206. [PMID: 36747655 PMCID: PMC9900797 DOI: 10.1101/2023.01.23.525206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The role of T cell help in autoantibody responses is not well understood. Since tolerance mechanisms govern both T and B cell responses, one might predict that both T cell tolerance and B cell tolerance must be defeated in autoantibody responses requiring T cell help. To define whether autoreactive B cells depend on T cells to generate autoantibody responses, we studied the role of T cells in autoantibody responses resulting from acute cell-specific deletion of regulatory phosphatases. Ars/A1 B cells are DNA-reactive and require continuous inhibitory signaling by the tyrosine phosphatase SHP-1 and the inositol phosphatases SHIP-1 and PTEN to maintain unresponsiveness. Acute B cell-restricted deletion of any of these phosphatases results in an autoantibody response. Here we show that CD40-CD40L interactions are required to support autoantibody responses of B cells whose anergy has been compromised. If the B cell-intrinsic driver of loss of tolerance is failed negative regulation of PI3K signaling, bystander T cells provide sufficient CD40-mediated signal 2 to support an autoantibody response. However, while autoantibody responses driven by acute B cell-targeted deletion of SHP-1 also require T cells, bystander T cell help does not suffice. These results demonstrate that upregulation of PI3K signaling in autoreactive B cells, recapitulating the effect of multiple autoimmunity risk alleles, promotes autoantibody responses both by increasing B cells’ cooperation with non-cognate T cell help, as well as by altering BCR signaling. Receptiveness to bystander T cell help enables autoreactive B cells to circumvent the fail-safe of T cell tolerance. Significance Phosphatase suppression of PI3K signaling is an important mechanism by which peripheral autoreactive B cells are kept in an unresponsive/anergic state. Loss of this suppression, due to genetic alleles that confer risk of autoimmunity, often occurs in autoreactive B cells of individuals who develop autoimmune disease. Here we demonstrate that de-repression of PI3K signaling promotes autoantibody responses of a DNA-reactive B cell clone by relaxing dependence of autoantibody responses on T cell-derived helper signals. These results suggest that impaired regulation of PI3K signaling can promote autoantibody responses in two ways: by restoring antigen receptor signaling and by enabling autoreactive B cells to circumvent restrictions imposed by T cell tolerance mechanisms.
Collapse
|
39
|
Wang C, Nistala R, Cao M, Li DP, Pan Y, Golzy M, Cui Y, Liu Z, Kang X. Repair of Limb Ischemia Is Dependent on Hematopoietic Stem Cell Specific-SHP-1 Regulation of TGF-β1. Arterioscler Thromb Vasc Biol 2023; 43:92-108. [PMID: 36412197 PMCID: PMC10037747 DOI: 10.1161/atvbaha.122.318205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Hematopoietic stem cell (HSC) therapy has shown promise for tissue regeneration after ischemia. Therefore, there is a need to understand mechanisms underlying endogenous HSCs activation in response to ischemic stress and coordination of angiogenesis and repair. SHP-1 plays important roles in HSC quiescence and differentiation by regulation of TGF-β1 signaling. TGF-β1 promotes angiogenesis by stimulating stem cells to secrete growth factors to initiate the formation of blood vessels and later aid in their maturation. We propose that SHP-1 responds to ischemia stress in HSC and progenitor cells (HSPC) via regulation of TGF-β1. METHODS A mouse hind limb ischemia model was used. Local blood perfusion in the limbs was determined using laser doppler perfusion imaging. The number of positive blood vessels per square millimeter, as well as blood vessel diameter (μm) and area (μm2), were calculated. Hematopoietic cells were analyzed using flow cytometry. The bone marrow transplantation assay was performed to measure HSC reconstitution. RESULTS After femoral artery ligation, TGF-β1 was initially decreased in the bone marrow by day 3 of ischemia, followed by an increase on day 7. This pattern was opposite to that in the peripheral blood, which is concordant with the response of HSC to ischemic stress. In contrast, SHP-1 deficiency in HSC is associated with irreversible activation of HSPCs in the bone marrow and increased circulating HSPCs in peripheral blood following limb ischemia. In addition, there was augmented auto-induction of TGF-β1 and sustained inactivation of SHP-1-Smad2 signaling, which impacted TGF-β1 expression in HSPCs in circulation. Importantly, restoration of normal T GF-β1 oscillations helped in the recovery of limb repair and function. CONCLUSIONS HSPC-SHP-1-mediated regulation of TGF-β1 in both bone marrow and peripheral blood is required for a normal response to ischemic stress.
Collapse
Affiliation(s)
- Chen Wang
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Ravi Nistala
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
- Division of Nephrology (R.N.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Min Cao
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - De-Pei Li
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Yi Pan
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Mojgan Golzy
- Department of Family and Community Medicine - Biostatistics Unit, School of Medicine, University of Missouri, Columbia (M.G.)
| | - Yuqi Cui
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
- Division of Cardiovascular Medicine (Y.C., Z.L.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - Zhenguo Liu
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
- Division of Cardiovascular Medicine (Y.C., Z.L.), Department of Medicine, University of Missouri School of Medicine, Columbia
| | - XunLei Kang
- Center for Precision Medicine (C.W., R.N., M.C., D.-P.L., Y.P., Y.C., Z.L., X.K.), Department of Medicine, University of Missouri School of Medicine, Columbia
| |
Collapse
|
40
|
Huse K, Bai B, Hilden VI, Bollum LK, Våtsveen TK, Munthe LA, Smeland EB, Irish JM, Wälchli S, Myklebust JH. Mechanism of CD79A and CD79B Support for IgM+ B Cell Fitness through B Cell Receptor Surface Expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2042-2053. [PMID: 36426942 PMCID: PMC9643646 DOI: 10.4049/jimmunol.2200144] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 09/09/2022] [Indexed: 12/31/2022]
Abstract
The BCR consists of surface-bound Ig and a heterodimeric signaling unit comprised of CD79A and CD79B. Upon cognate Ag recognition, the receptor initiates important signals for B cell development and function. The receptor also conveys Ag-independent survival signals termed tonic signaling. Although the requirement of a CD79A/CD79B heterodimer for BCR complex assembly and surface expression is well established based on mice models, few studies have investigated this in human mature B cells. In this study, we found that human tonsillar B cells with high surface expression of IgM or IgG had potentiated BCR signaling compared with BCRlow cells, and high IgM expression in germinal center B cells was associated with reduced apoptosis. We explored the mechanism for IgM surface expression by CRISPR/Cas9-induced deletion of CD79A or CD79B in four B lymphoma cell lines. Deletion of either CD79 protein caused loss of surface IgM in all cell lines and reduced fitness in three. From two cell lines, we generated stable CD79A or CD79B knockout clones and demonstrated that loss of CD79A or CD79B caused a block in N-glycan maturation and accumulation of immature proteins, compatible with retention of BCR components in the endoplasmic reticulum. Rescue experiments with CD79B wild-type restored surface expression of CD79A and IgM with mature glycosylation, whereas a naturally occurring CD79B G137S mutant disrupting CD79A/CD79B heterodimerization did not. Our study highlights that CD79A and CD79B are required for surface IgM expression in human B cells and illuminates the importance of the IgM expression level for signaling and fitness.
Collapse
Affiliation(s)
- Kanutte Huse
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Baoyan Bai
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Norway
| | - Vera Irene Hilden
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lise K Bollum
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thea K Våtsveen
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Div. of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Ludvig A Munthe
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Div. of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Erlend B Smeland
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jonathan Michael Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Cancer Treatment, Oslo University Hospital, Oslo, Norway
| | - June H. Myklebust
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B-cell malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
Guerau-de-Arellano M, Piedra-Quintero ZL, Tsichlis PN. Akt isoforms in the immune system. Front Immunol 2022; 13:990874. [PMID: 36081513 PMCID: PMC9445622 DOI: 10.3389/fimmu.2022.990874] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Akt is a PI3K-activated serine-threonine kinase that exists in three distinct isoforms. Akt's expression in most immune cells, either at baseline or upon activation, reflects its importance in the immune system. While Akt is most highly expressed in innate immune cells, it plays crucial roles in both innate and adaptive immune cell development and/or effector functions. In this review, we explore what's known about the role of Akt in innate and adaptive immune cells. Wherever possible, we discuss the overlapping and distinct role of the three Akt isoforms, namely Akt1, Akt2, and Akt3, in immune cells.
Collapse
Affiliation(s)
- Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States,Department of Neuroscience, The Ohio State University, Columbus, OH, United States,The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States,*Correspondence: Mireia Guerau-de-Arellano,
| | - Zayda L. Piedra-Quintero
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Philip N. Tsichlis
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States,Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
42
|
Gammaherpesvirus-mediated repression reveals EWSR1 to be a negative regulator of B cell responses. Proc Natl Acad Sci U S A 2022; 119:e2123362119. [PMID: 35921433 PMCID: PMC9371696 DOI: 10.1073/pnas.2123362119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The germinal center (GC) plays a central role in the generation of antigen-specific B cells and antibodies. Tight regulation of the GC is essential due to the inherent risks of tumorigenesis and autoimmunity posed by inappropriate GC B cell processes. Gammaherpesviruses such as Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) utilize numerous armaments to drive infected naïve B cells, independent of antigen, through GC reactions to expand the latently infected B cell population and establish a stable latency reservoir. We previously demonstrated that the MHV68 microRNA (miRNA) mghv-miR-M1-7-5p represses host EWSR1 (Ewing sarcoma breakpoint region 1) to promote B cell infection. EWSR1 is a transcription and splicing regulator that is recognized for its involvement as a fusion protein in Ewing sarcoma. A function for EWSR1 in B cell responses has not been previously reported. Here, we demonstrate that 1) B cell-specific deletion of EWSR1 had no effect on generation of mature B cell subsets or basal immunoglobulin levels in naïve mice, 2) repression or ablation of EWSR1 in B cells promoted expansion of MHV68 latently infected GC B cells, and 3) B cell-specific deletion of EWSR1 during a normal immune response to nonviral antigen resulted in significantly elevated numbers of antigen-specific GC B cells, plasma cells, and circulating antibodies. Notably, EWSR1 deficiency did not affect the proliferation or survival of GC B cells but instead resulted in the generation of increased numbers of precursor GC B cells. Cumulatively, these findings demonstrate that EWSR1 is a negative regulator of B cell responses.
Collapse
|
43
|
Dvorscek AR, McKenzie CI, Robinson MJ, Ding Z, Pitt C, O'Donnell K, Zotos D, Brink R, Tarlinton DM, Quast I. IL-21 has a critical role in establishing germinal centers by amplifying early B cell proliferation. EMBO Rep 2022; 23:e54677. [PMID: 35801309 PMCID: PMC9442303 DOI: 10.15252/embr.202254677] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022] Open
Abstract
The proliferation and differentiation of antigen‐specific B cells, including the generation of germinal centers (GC), are prerequisites for long‐lasting, antibody‐mediated immune protection. Affinity for antigen determines B cell recruitment, proliferation, differentiation, and competitiveness in the response, largely through determining access to T cell help. However, how T cell‐derived signals contribute to these outcomes is incompletely understood. Here, we report how the signature cytokine of follicular helper T cells, IL‐21, acts as a key regulator of the initial B cell response by accelerating cell cycle progression and the rate of cycle entry, increasing their contribution to the ensuing GC. This effect occurs over a wide range of initial B cell receptor affinities and correlates with elevated AKT and S6 phosphorylation. Moreover, the resultant increased proliferation can explain the IL‐21‐mediated promotion of plasma cell differentiation. Collectively, our data establish that IL‐21 acts from the outset of a T cell‐dependent immune response to increase cell cycle progression and fuel cyclic re‐entry of B cells, thereby regulating the initial GC size and early plasma cell output.
Collapse
Affiliation(s)
- Alexandra R Dvorscek
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Craig I McKenzie
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Marcus J Robinson
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Zhoujie Ding
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Catherine Pitt
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Kristy O'Donnell
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Dimitra Zotos
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Robert Brink
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
| | - David M Tarlinton
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| | - Isaak Quast
- Department of Immunology and Pathology, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
44
|
Krollmann C, Cieslak K, Koerber RM, Luksch H, Rösen-Wolff A, Brossart P, Teichmann LL. Quantification of unperturbed phosphoprotein levels in immune cell subsets with phosphoflow to assess immune signaling in autoimmune disease. STAR Protoc 2022; 3:101309. [PMID: 35496781 PMCID: PMC9038771 DOI: 10.1016/j.xpro.2022.101309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
45
|
Vaidehi Narayanan H, Hoffmann A. From Antibody Repertoires to Cell-Cell Interactions to Molecular Networks: Bridging Scales in the Germinal Center. Front Immunol 2022; 13:898078. [PMID: 35603162 PMCID: PMC9114758 DOI: 10.3389/fimmu.2022.898078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/08/2022] [Indexed: 01/02/2023] Open
Abstract
Antibody-mediated adaptive immunity must provide effective long-term protection with minimal adverse effects, against rapidly mutating pathogens, in a human population with diverse ages, genetics, and immune histories. In order to grasp and leverage the complexities of the antibody response, we advocate for a mechanistic understanding of the multiscale germinal center (GC) reaction - the process by which precursor B-cells evolve high-affinity antigen-specific antibodies, forming an effector repertoire of plasma and memory cells for decades-long protection. The regulatory dynamics of B-cells within the GC are complex, and unfold across multiple interacting spatial and temporal scales. At the organism scale, over weeks to years, the antibody sequence repertoire formed by various B-cell clonal lineages modulates antibody quantity and quality over time. At the tissue and cellular scale, over hours to weeks, B-cells undergo selection via spatially distributed interactions with local stroma, antigen, and helper T-cells. At the molecular scale, over seconds to days, intracellular signaling, transcriptional, and epigenetic networks modulate B-cell fates and shape their clonal lineages. We summarize our current understanding within each of these scales, and identify missing links in connecting them. We suggest that quantitative multi-scale mathematical models of B-cell and GC reaction dynamics provide predictive frameworks that can apply basic immunological knowledge to practical challenges such as rational vaccine design.
Collapse
Affiliation(s)
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, and Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
46
|
Enterina JR, Sarkar S, Streith L, Jung J, Arlian BM, Meyer SJ, Takematsu H, Xiao C, Baldwin TA, Nitschke L, Shlomchick MJ, Paulson JC, Macauley MS. Coordinated changes in glycosylation regulate the germinal center through CD22. Cell Rep 2022; 38:110512. [PMID: 35294874 PMCID: PMC9018098 DOI: 10.1016/j.celrep.2022.110512] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/19/2022] [Accepted: 02/16/2022] [Indexed: 12/18/2022] Open
Abstract
Germinal centers (GCs) are essential for antibody affinity maturation. GC B cells have a unique repertoire of cell surface glycans compared with naive B cells, yet functional roles for changes in glycosylation in the GC have yet to be ascribed. Detection of GCs by the antibody GL7 reflects a downregulation in ligands for CD22, an inhibitory co-receptor of the B cell receptor. To test a functional role for downregulation of CD22 ligands in the GC, we generate a mouse model that maintains CD22 ligands on GC B cells. With this model, we demonstrate that glycan remodeling plays a critical role in the maintenance of B cells in the GC. Sustained expression of CD22 ligands induces higher levels of apoptosis in GC B cells, reduces memory B cell and plasma cell output, and delays affinity maturation of antibodies. These defects are CD22 dependent, demonstrating that downregulation of CD22 ligands on B cells plays a critical function in the GC.
Collapse
Affiliation(s)
- Jhon R Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Laura Streith
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sarah J Meyer
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Hiromu Takematsu
- Faculty of Medical Technology, Fujita Health University, Aichi 470-1192, Japan
| | - Changchun Xiao
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Mark J Shlomchick
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Matthew S Macauley
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada; Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
47
|
Abstract
Germinal centers (GCs) are microanatomical sites of B cell clonal expansion and antibody affinity maturation. Therein, B cells undergo the Darwinian process of somatic diversification and affinity-driven selection of immunoglobulins that produces the high-affinity antibodies essential for effective humoral immunity. Here, we review recent developments in the field of GC biology, primarily as it pertains to GCs induced by infection or immunization. First, we summarize the phenotype and function of the different cell types that compose the GC, focusing on GC B cells. Then, we review the cellular and molecular bases of affinity-dependent selection within the GC and the export of memory and plasma cells. Finally, we present an overview of the emerging field of GC clonal dynamics, focusing on how GC and post-GC selection shapes the diversity of antibodies secreted into serum. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA;
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology and Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA;
| |
Collapse
|
48
|
Zotos D, Quast I, Li-Wai-Suen CSN, McKenzie CI, Robinson MJ, Kan A, Smyth GK, Hodgkin PD, Tarlinton DM. The concerted change in the distribution of cell cycle phases and zone composition in germinal centers is regulated by IL-21. Nat Commun 2021; 12:7160. [PMID: 34887406 PMCID: PMC8660905 DOI: 10.1038/s41467-021-27477-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Humoral immune responses require germinal centres (GC) for antibody affinity maturation. Within GC, B cell proliferation and mutation are segregated from affinity-based positive selection in the dark zone (DZ) and light zone (LZ) substructures, respectively. While IL-21 is known to be important in affinity maturation and GC maintenance, here we show it is required for both establishing normal zone representation and preventing the accumulation of cells in the G1 cell cycle stage in the GC LZ. Cell cycle progression of DZ B cells is unaffected by IL-21 availability, as is the zone phenotype of the most highly proliferative GC B cells. Collectively, this study characterises the development of GC zones as a function of time and B cell proliferation and identifies IL-21 as an important regulator of these processes. These data help explain the requirement for IL-21 in normal antibody affinity maturation.
Collapse
Affiliation(s)
- Dimitra Zotos
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Isaak Quast
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Connie S N Li-Wai-Suen
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melboure, Parkville, VIC, 3010, Australia
| | - Craig I McKenzie
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Marcus J Robinson
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Andrey Kan
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- School of Computer Science, University of Adelaide, Frome Rd, Adelaide, SA, 5005, Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- School of Mathematics and Statistics, University of Melboure, Parkville, VIC, 3010, Australia
| | - Philip D Hodgkin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - David M Tarlinton
- Department of Immunology and Pathology, Monash University, 89 Commercial Road, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
49
|
Gaudette BT, Roman CJ, Ochoa TA, Gómez Atria D, Jones DD, Siebel CW, Maillard I, Allman D. Resting innate-like B cells leverage sustained Notch2/mTORC1 signaling to achieve rapid and mitosis-independent plasma cell differentiation. J Clin Invest 2021; 131:e151975. [PMID: 34473651 DOI: 10.1172/jci151975] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Little is known about how cells regulate and integrate distinct biosynthetic pathways governing differentiation and cell division. For B lineage cells it is widely accepted that activated cells must complete several rounds of mitosis before yielding antibody-secreting plasma cells. However, we report that marginal zone (MZ) B cells, innate-like naive B cells known to generate plasma cells rapidly in response to blood-borne bacteria, generate functional plasma cells despite cell-cycle arrest. Further, short-term Notch2 blockade in vivo reversed division-independent differentiation potential and decreased transcript abundance for numerous mTORC1- and Myc-regulated genes. Myc loss compromised plasma cell differentiation for MZ B cells, and reciprocally induced ectopic mTORC1 signaling in follicular B cells enabled division-independent differentiation and plasma cell-affiliated gene expression. We conclude that ongoing in situ Notch2/mTORC1 signaling in MZ B cells establishes a unique cellular state that enables rapid division-independent plasma cell differentiation.
Collapse
Affiliation(s)
| | - Carly J Roman
- The Department of Pathology and Laboratory Medicine and
| | - Trini A Ochoa
- The Department of Pathology and Laboratory Medicine and
| | - Daniela Gómez Atria
- The Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Derek D Jones
- The Department of Pathology and Laboratory Medicine and
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech Inc., South San Francisco, California, USA
| | - Ivan Maillard
- The Department of Medicine, Division of Hematology/Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Allman
- The Department of Pathology and Laboratory Medicine and
| |
Collapse
|
50
|
Verstegen NJM, Ubels V, Westerhoff HV, van Ham SM, Barberis M. System-Level Scenarios for the Elucidation of T Cell-Mediated Germinal Center B Cell Differentiation. Front Immunol 2021; 12:734282. [PMID: 34616402 PMCID: PMC8488341 DOI: 10.3389/fimmu.2021.734282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Germinal center (GC) reactions are vital to the correct functioning of the adaptive immune system, through formation of high affinity, class switched antibodies. GCs are transient anatomical structures in secondary lymphoid organs where specific B cells, after recognition of antigen and with T cell help, undergo class switching. Subsequently, B cells cycle between zones of proliferation and somatic hypermutation and zones where renewed antigen acquisition and T cell help allows for selection of high affinity B cells (affinity maturation). Eventually GC B cells first differentiate into long-lived memory B cells (MBC) and finally into plasma cells (PC) that partially migrate to the bone marrow to encapsulate into long-lived survival niches. The regulation of GC reactions is a highly dynamically coordinated process that occurs between various cells and molecules that change in their signals. Here, we present a system-level perspective of T cell-mediated GC B cell differentiation, presenting and discussing the experimental and computational efforts on the regulation of the GCs. We aim to integrate Systems Biology with B cell biology, to advance elucidation of the regulation of high-affinity, class switched antibody formation, thus to shed light on the delicate functioning of the adaptive immune system. Specifically, we: i) review experimental findings of internal and external factors driving various GC dynamics, such as GC initiation, maturation and GCBC fate determination; ii) draw comparisons between experimental observations and mathematical modeling investigations; and iii) discuss and reflect on current strategies of modeling efforts, to elucidate B cell behavior during the GC tract. Finally, perspectives are specifically given on to the areas where a Systems Biology approach may be useful to predict novel GCBC-T cell interaction dynamics.
Collapse
Affiliation(s)
- Niels J M Verstegen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Victor Ubels
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford, United Kingdom
| | - Hans V Westerhoff
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands.,Department of Molecular Cell Physiology, VU University Amsterdam, Amsterdam, Netherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands.,Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford, United Kingdom
| |
Collapse
|