Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.304
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 39: 345-368, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33556247

ABSTRACT

For many infections and almost all vaccines, neutralizing-antibody-mediated immunity is the primary basis and best functional correlate of immunological protection. Durable long-term humoral immunity is mediated by antibodies secreted by plasma cells that preexist subsequent exposures and by memory B cells that rapidly respond to infections once they have occurred. In the midst of the current pandemic of coronavirus disease 2019, it is important to define our current understanding of the unique roles of memory B cells and plasma cells in immunity and the factors that control the formation and persistence of these cell types. This fundamental knowledge is the basis to interpret findings from natural infections and vaccines. Here, we review transcriptional and metabolic programs that promote and support B cell fates and functions, suggesting points at which these pathways do and do not intersect.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Energy Metabolism , Gene Expression Regulation , Immunologic Memory , Plasma Cells/immunology , Plasma Cells/metabolism , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Germinal Center/immunology , Germinal Center/metabolism , Humans , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Transcription, Genetic
2.
Annu Rev Immunol ; 36: 339-357, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29356584

ABSTRACT

Maintenance of immunological self-tolerance requires lymphocytes carrying self-reactive antigen receptors to be selectively prevented from mounting destructive or inflammatory effector responses. Classically, self-tolerance is viewed in terms of the removal, editing, or silencing of B and T cells that have formed self-reactive antigen receptors during their early development. However, B cells activated by foreign antigen can enter germinal centers (GCs), where they further modify their antigen receptor by somatic hypermutation (SHM) of their immunoglobulin genes. The inevitable emergence of activated, self-reactive GC B cells presents a unique challenge to the maintenance of self-tolerance that must be rapidly countered to avoid autoantibody production. Here we discuss current knowledge of the mechanisms that enforce B cell self-tolerance, with particular focus on the control of self-reactive GC B cells. We also consider how self-reactive GC B cells can escape self-tolerance to initiate autoantibody production or instead be redeemed via SHM and used in productive antibody responses.


Subject(s)
Autoimmunity , B-Lymphocytes/immunology , Germinal Center/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/metabolism , Germinal Center/metabolism , Humans , Immune Tolerance , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
3.
Nat Immunol ; 25(6): 1097-1109, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38698087

ABSTRACT

Affinity-matured plasma cells (PCs) of varying lifespans are generated through a germinal center (GC) response. The developmental dynamics and genomic programs of antigen-specific PC precursors remain to be elucidated. Here, using a model antigen in mice, we demonstrate biphasic generation of PC precursors, with those generating long-lived bone marrow PCs preferentially produced in the late phase of GC response. Clonal tracing using single-cell RNA sequencing and B cell antigen receptor sequencing in spleen and bone marrow compartments, coupled with adoptive transfer experiments, reveals a new PC transition state that gives rise to functionally competent PC precursors. The latter undergo clonal expansion, dependent on inducible expression of TIGIT. We propose a model for the proliferation and programming of precursors of long-lived PCs, based on extended antigen encounters in the GC.


Subject(s)
Cell Differentiation , Germinal Center , Plasma Cells , Animals , Plasma Cells/immunology , Plasma Cells/metabolism , Mice , Germinal Center/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/genetics , Mice, Inbred C57BL , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Mice, Transgenic
4.
Nat Immunol ; 25(7): 1283-1295, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38862796

ABSTRACT

While some infections elicit germinal centers, others produce only extrafollicular responses. The mechanisms controlling these dichotomous fates are poorly understood. We identify IL-12 as a cytokine switch, acting directly on B cells to promote extrafollicular and suppress germinal center responses. IL-12 initiates a B cell-intrinsic feed-forward loop between IL-12 and IFNγ, amplifying IFNγ production, which promotes proliferation and plasmablast differentiation from mouse and human B cells, in synergy with IL-12. IL-12 sustains the expression of a portion of IFNγ-inducible genes. Together, they also induce unique gene changes, reflecting both IFNγ amplification and cooperative effects between both cytokines. In vivo, cells lacking both IL-12 and IFNγ receptors are more impaired in plasmablast production than those lacking either receptor alone. Further, B cell-derived IL-12 enhances both plasmablast responses and T helper 1 cell commitment. Thus, B cell-derived IL-12, acting on T and B cells, determines the immune response mode, with implications for vaccines, pathogen protection and autoimmunity.


Subject(s)
B-Lymphocytes , Cell Differentiation , Germinal Center , Interferon-gamma , Interleukin-12 , Animals , Interleukin-12/immunology , Interleukin-12/metabolism , Mice , Interferon-gamma/metabolism , Interferon-gamma/immunology , Germinal Center/immunology , Humans , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Mice, Knockout , Mice, Inbred C57BL , Plasma Cells/immunology , Plasma Cells/metabolism , Lymphocyte Activation/immunology , Receptors, Interferon/metabolism , Receptors, Interferon/genetics , Cells, Cultured , Cell Proliferation
5.
Cell ; 184(7): 1775-1789.e19, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33711260

ABSTRACT

Regulatory T cells prevent the emergence of autoantibodies and excessive IgE, but the precise mechanisms are unclear. Here, we show that BCL6-expressing Tregs, known as follicular regulatory T (Tfr) cells, produce abundant neuritin protein that targets B cells. Mice lacking Tfr cells or neuritin in Foxp3-expressing cells accumulated early plasma cells in germinal centers (GCs) and developed autoantibodies against histones and tissue-specific self-antigens. Upon immunization, these mice also produced increased plasma IgE and IgG1. We show that neuritin is taken up by B cells, causes phosphorylation of numerous proteins, and dampens IgE class switching. Neuritin reduced differentiation of mouse and human GC B cells into plasma cells, downregulated BLIMP-1, and upregulated BCL6. Administration of neuritin to Tfr-deficient mice prevented the accumulation of early plasma cells in GCs. Production of neuritin by Tfr cells emerges as a central mechanism to suppress B cell-driven autoimmunity and IgE-mediated allergies.


Subject(s)
B-Lymphocytes/immunology , Nerve Tissue Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoantibodies/immunology , Autoimmunity , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , GPI-Linked Proteins/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Histones/immunology , Immunoglobulin Class Switching , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
6.
Cell ; 179(1): 3-7, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31519306

ABSTRACT

This year's Lasker Basic Medical Research Award honors Max Cooper and Jacques Miller for discoveries that revealed the organizing principles of adaptive immunity. Their collective contributions have had broad clinical impact in the treatment of immune disease.


Subject(s)
Adaptive Immunity/immunology , Cell Communication/immunology , Plasma Cells/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation/immunology , Antigen Presentation , Bone Marrow/immunology , Chickens , Humans , Hybridomas/immunology , Immunoglobulin Class Switching/immunology , Lymph Nodes/immunology , Mice , Nobel Prize , Self Tolerance/immunology , Thymus Gland/immunology
7.
Cell ; 177(3): 524-540, 2019 04 18.
Article in English | MEDLINE | ID: mdl-31002794

ABSTRACT

B cells and the antibodies they produce have a deeply penetrating influence on human physiology. Here, we review current understanding of how B cell responses are initiated; the different paths to generate short- and long-lived plasma cells, germinal center cells, and memory cells; and how each path impacts antibody diversity, selectivity, and affinity. We discuss how basic research is informing efforts to generate vaccines that induce broadly neutralizing antibodies against viral pathogens, revealing the special features associated with allergen-reactive IgE responses and uncovering the antibody-independent mechanisms by which B cells contribute to health and disease.


Subject(s)
B-Lymphocytes/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/metabolism , Antigens/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , Germinal Center/metabolism , Humans , Immunologic Memory , Plasma Cells/immunology , Plasma Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vaccines/immunology
8.
Nat Immunol ; 21(7): 790-801, 2020 07.
Article in English | MEDLINE | ID: mdl-32424361

ABSTRACT

Plasmodium parasite-specific antibodies are critical for protection against malaria, yet the development of long-lived and effective humoral immunity against Plasmodium takes many years and multiple rounds of infection and cure. Here, we report that the rapid development of short-lived plasmablasts during experimental malaria unexpectedly hindered parasite control by impeding germinal center responses. Metabolic hyperactivity of plasmablasts resulted in nutrient deprivation of the germinal center reaction, limiting the generation of memory B cell and long-lived plasma cell responses. Therapeutic administration of a single amino acid to experimentally infected mice was sufficient to overcome the metabolic constraints imposed by plasmablasts and enhanced parasite clearance and the formation of protective humoral immune memory responses. Thus, our studies not only challenge the current model describing the role and function of blood-stage Plasmodium-induced plasmablasts but they also reveal new targets and strategies to improve anti-Plasmodium humoral immunity.


Subject(s)
Immunity, Humoral , Malaria/immunology , Plasma Cells/metabolism , Plasmodium falciparum/immunology , Adolescent , Adult , Amino Acids/administration & dosage , Amino Acids/metabolism , Animals , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antibodies, Protozoan/metabolism , Antimalarials/administration & dosage , DNA, Protozoan/isolation & purification , Disease Models, Animal , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Host-Parasite Interactions/immunology , Humans , Malaria/blood , Malaria/drug therapy , Malaria/parasitology , Mice , Mice, Transgenic , Middle Aged , Nutrients/metabolism , Plasma Cells/immunology , Plasma Cells/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Proof of Concept Study , Young Adult
9.
Nat Immunol ; 20(1): 97-108, 2019 01.
Article in English | MEDLINE | ID: mdl-30510223

ABSTRACT

Memory B cells are found in lymphoid and non-lymphoid tissues, suggesting that some may be tissue-resident cells. Here we show that pulmonary influenza infection elicited lung-resident memory B cells (BRM cells) that were phenotypically and functionally distinct from their systemic counterparts. BRM cells were established in the lung early after infection, in part because their placement required local antigen encounter. Lung BRM cells, but not systemic memory B cells, contributed to early plasmablast responses following challenge infection. Following secondary infection, antigen-specific BRM cells differentiated in situ, whereas antigen-non-specific BRM cells were maintained as memory cells. These data demonstrate that BRM cells are an important component of immunity to respiratory viruses such as influenza virus and suggest that vaccines designed to elicit BRM cells must deliver antigen to the lungs.


Subject(s)
Antigens, Viral/immunology , B-Lymphocytes/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/physiology , Plasma Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Female , Humans , Immunity, Humoral , Immunologic Memory , Lung/virology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
Nat Immunol ; 19(9): 1025-1034, 2018 09.
Article in English | MEDLINE | ID: mdl-30082831

ABSTRACT

Memory B cells (Bmem cells) are the basis of long-lasting humoral immunity. They respond to re-encountered antigens by rapidly producing specific antibodies and forming germinal centers (GCs), a recall response that has been known for decades but remains poorly understood. We found that the receptor for the cytokine IL-9 (IL-9R) was induced selectively on Bmem cells after primary immunization and that IL-9R-deficient mice exhibited a normal primary antibody response but impaired recall antibody responses, with attenuated population expansion and plasma-cell differentiation of Bmem cells. In contrast, there was augmented GC formation, possibly due to defective downregulation of the ligand for the co-stimulatory receptor ICOS on Bmem cells. A fraction of Bmem cells produced IL-9. These findings indicate that IL-9R signaling in Bmem cells regulates humoral recall responses.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/physiology , Interleukin-9/metabolism , Plasma Cells/immunology , Receptors, Interleukin-9/genetics , Animals , Cell Differentiation , Cells, Cultured , Immunity, Humoral , Immunization, Secondary , Immunoglobulin Variable Region/genetics , Immunologic Memory , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-9/metabolism , Signal Transduction
11.
Immunity ; 54(9): 2005-2023.e10, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525339

ABSTRACT

Cell fate decisions during early B cell activation determine the outcome of responses to pathogens and vaccines. We examined the early B cell response to T-dependent antigen in mice by single-cell RNA sequencing. Early after immunization, a homogeneous population of activated precursors (APs) gave rise to a transient wave of plasmablasts (PBs), followed a day later by the emergence of germinal center B cells (GCBCs). Most APs rapidly exited the cell cycle, giving rise to non-GC-derived early memory B cells (eMBCs) that retained an AP-like transcriptional profile. Rapid decline of antigen availability controlled these events; provision of excess antigen precluded cell cycle exit and induced a new wave of PBs. Fate mapping revealed a prominent contribution of eMBCs to the MBC pool. Quiescent cells with an MBC phenotype dominated the early response to immunization in primates. A reservoir of APs/eMBCs may enable rapid readjustment of the immune response when failure to contain a threat is manifested by increased antigen availability.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Immunity, Humoral/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , Antigen Presentation/immunology , Cell Differentiation/immunology , Mice , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology
12.
Immunity ; 54(12): 2772-2783.e5, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788602

ABSTRACT

Humoral immunity is essential for protection against pathogens, emphasized by the prevention of 2-3 million deaths worldwide annually by childhood immunizations. Long-term protective immunity is dependent on the continual production of neutralizing antibodies by the subset of long-lived plasma cells (LLPCs). LLPCs are not intrinsically long-lived, but require interaction with LLPC niche stromal cells for survival. However, it remains unclear which and how these interactions sustain LLPC survival and long-term humoral immunity. We now have found that the immunosuppressive enzyme indoleamine 2,3- dioxygenase 1 (IDO1) is required to sustain antibody responses and LLPC survival. Activation of IDO1 occurs upon the engagement of CD80/CD86 on the niche dendritic cells by CD28 on LLPC. Kynurenine, the product of IDO1 catabolism, activates the aryl hydrocarbon receptor in LLPC, reinforcing CD28 expression and survival signaling. These findings expand the immune function of IDO1 and uncover a novel pathway for sustaining LLPC survival and humoral immunity.


Subject(s)
Dendritic Cells/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Plasma Cells/immunology , Animals , Antibodies, Neutralizing/metabolism , B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Cell Self Renewal , Cell Survival , Cells, Cultured , Female , Immunity, Humoral , Immunologic Memory , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mice , Mice, Knockout
13.
Immunity ; 54(2): 235-246.e5, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33357409

ABSTRACT

The interleukin-6 (IL-6) membrane receptor and its circulating soluble form, sIL-6R, can be targeted by antibody therapy to reduce deleterious immune signaling caused by chronic overexpression of the pro-inflammatory cytokine IL-6. This strategy may also hold promise for treating acute hyperinflammation, such as observed in coronavirus disease 2019 (COVID-19), highlighting a need to define regulators of IL-6 homeostasis. We found that conventional dendritic cells (cDCs), defined in mice via expression of the transcription factor Zbtb46, were a major source of circulating sIL-6R and, thus, systemically regulated IL-6 signaling. This was uncovered through identification of a cDC-dependent but T cell-independent modality that naturally adjuvants plasma cell differentiation and antibody responses to protein antigens. This pathway was then revealed as part of a broader biological buffer system in which cDC-derived sIL-6R set the in-solution persistence of IL-6. This control axis may further inform the development of therapeutic agents to modulate pro-inflammatory immune reactions.


Subject(s)
Dendritic Cells/immunology , Interleukin-6/blood , Interleukin-6/immunology , ADAM17 Protein , Animals , Cell Differentiation , Immunity, Humoral , Immunoglobulin M/immunology , Inflammation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interleukin-6/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/immunology , Receptors, Interleukin-6/blood , Receptors, Interleukin-6/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 7/immunology
14.
Immunity ; 54(9): 2024-2041.e8, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34473957

ABSTRACT

Sepsis results in elevated adenosine in circulation. Extracellular adenosine triggers immunosuppressive signaling via the A2a receptor (A2aR). Sepsis survivors develop persistent immunosuppression with increased risk of recurrent infections. We utilized the cecal ligation and puncture (CLP) model of sepsis and subsequent infection to assess the role of adenosine in post-sepsis immune suppression. A2aR-deficient mice showed improved resistance to post-sepsis infections. Sepsis expanded a subset of CD39hi B cells and elevated extracellular adenosine, which was absent in mice lacking CD39-expressing B cells. Sepsis-surviving B cell-deficient mice were more resistant to secondary infections. Mechanistically, metabolic reprogramming of septic B cells increased production of ATP, which was converted into adenosine by CD39 on plasmablasts. Adenosine signaling via A2aR impaired macrophage bactericidal activity and enhanced interleukin-10 production. Septic individuals exhibited expanded CD39hi plasmablasts and adenosine accumulation. Our study reveals CD39hi plasmablasts and adenosine as important drivers of sepsis-induced immunosuppression with relevance in human disease.


Subject(s)
Adenosine/immunology , Antigens, CD/immunology , Apyrase/immunology , Immune Tolerance/immunology , Macrophages/immunology , Plasma Cells/immunology , Sepsis/immunology , Adenosine/metabolism , Animals , Antigens, CD/metabolism , Apyrase/metabolism , Cellular Reprogramming/immunology , Macrophages/metabolism , Mice , Plasma Cells/metabolism , Receptor, Adenosine A2A/immunology , Receptor, Adenosine A2A/metabolism , Sepsis/metabolism
15.
Immunity ; 54(10): 2273-2287.e6, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644558

ABSTRACT

Diets high in cholesterol alter intestinal immunity. Here, we examined how the cholesterol metabolite 25-hydroxycholesterol (25-HC) impacts the intestinal B cell response. Mice lacking cholesterol 25-hydroxylase (CH25H), the enzyme generating 25-HC, had higher frequencies of immunoglobulin A (IgA)-secreting antigen-specific B cells upon immunization or infection. 25-HC did not affect class-switch recombination but rather restrained plasma cell (PC) differentiation. 25-HC was produced by follicular dendritic cells and increased in response to dietary cholesterol. Mechanistically, 25-HC restricted activation of the sterol-sensing transcription factor SREBP2, thereby regulating B cell cholesterol biosynthesis. Ectopic expression of SREBP2 in germinal center B cells induced rapid PC differentiation, whereas SREBP2 deficiency reduced PC output in vitro and in vivo. High-cholesterol diet impaired, whereas Ch25h deficiency enhanced, the IgA response against Salmonella and the resulting protection from systemic bacterial dissemination. Thus, a 25-HC-SREBP2 axis shapes the humoral response at the intestinal barrier, providing insight into the effect of high dietary cholesterol in intestinal immunity.


Subject(s)
Cell Differentiation/immunology , Hydroxycholesterols/metabolism , Immunoglobulin A/immunology , Plasma Cells/immunology , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Cholesterol, Dietary/immunology , Cholesterol, Dietary/metabolism , Hydroxycholesterols/immunology , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Peyer's Patches/immunology , Peyer's Patches/metabolism , Plasma Cells/metabolism
16.
Immunity ; 54(12): 2784-2794.e6, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34626548

ABSTRACT

Self-reactive B cell progenitors are eliminated through central tolerance checkpoints, a process thought to be restricted to the bone marrow in mammals. Here, we identified a consecutive trajectory of B cell development in the meninges of mice and non-human primates. The meningeal B cells were located predominantly at the dural sinuses, where endothelial cells expressed essential niche factors to support B cell development. Parabiosis experiments together with lineage tracing showed that meningeal developing B cells were replenished continuously from hematopoietic stem cell (HSC)-derived progenitors via a circulation-independent route. Autoreactive immature B cells that recognized myelin oligodendrocyte glycoprotein (MOG), a central nervous system-specific antigen, were eliminated specifically from the meninges. Furthermore, genetic deletion of the Mog gene restored the self-reactive B cell population in the meninges. These findings identify the meninges as a distinct reservoir for B cell development, allowing in situ negative selection to ensure a locally non-self-reactive immune repertoire.


Subject(s)
Dendritic Cells/immunology , Hematopoietic Stem Cells/physiology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Meninges/immunology , Plasma Cells/immunology , Animals , Antibodies, Neutralizing/metabolism , B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Cell Self Renewal , Cell Survival , Cells, Cultured , Humans , Immunity, Humoral , Immunologic Memory , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Mice , Mice, Inbred C57BL
17.
Immunity ; 54(2): 205-210, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33513337

ABSTRACT

Immunological memory is a mechanism to protect us against reinfection. Antibodies produced by B cells are integral to this defense strategy and underlie virtually all vaccine success. Here, we explain how B cells memory is generated by infection and vaccination, what influences its efficacy and its persistence, and how characterizing these parameters in the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) will help achieve protective immunity through vaccination.


Subject(s)
B-Lymphocytes/immunology , COVID-19/immunology , Immunologic Memory , SARS-CoV-2/immunology , Antibodies, Viral/immunology , COVID-19/prevention & control , Humans , Plasma Cells/immunology , T-Lymphocytes/immunology , Vaccination , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
18.
Immunity ; 54(12): 2756-2771.e10, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34879220

ABSTRACT

In contrast to other antibody isotypes, B cells switched to IgE respond transiently and do not give rise to long-lived plasma cells (PCs) or memory B cells. To better understand IgE-BCR-mediated control of IgE responses, we developed whole-genome CRISPR screening that enabled comparison of IgE+ and IgG1+ B cell requirements for proliferation, survival, and differentiation into PCs. IgE+ PCs exhibited dependency on the PI3K-mTOR axis that increased protein amounts of the transcription factor IRF4. In contrast, loss of components of the calcium-calcineurin-NFAT pathway promoted IgE+ PC differentiation. Mice bearing a B cell-specific deletion of calcineurin B1 exhibited increased production of IgE+ PCs. Mechanistically, sustained elevation of intracellular calcium in IgE+ PCs downstream of the IgE-BCR promoted BCL2L11-dependent apoptosis. Thus, chronic calcium signaling downstream of the IgE-BCR controls the self-limiting character of IgE responses and may be relevant to the accumulation of IgE-producing cells in allergic disease.


Subject(s)
B-Lymphocyte Subsets/immunology , Calcineurin/metabolism , Hypersensitivity/immunology , Plasma Cells/immunology , Animals , Apoptosis , Bcl-2-Like Protein 11/metabolism , Calcineurin/genetics , Calcium Signaling , Cell Differentiation , Cell Survival , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Humans , Immunoglobulin E/metabolism , Immunoglobulin G/metabolism , Immunologic Memory , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism
19.
Immunity ; 54(5): 1083-1095.e7, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33891889

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) is a life-threatening post-infectious complication occurring unpredictably weeks after mild or asymptomatic SARS-CoV-2 infection. We profiled MIS-C, adult COVID-19, and healthy pediatric and adult individuals using single-cell RNA sequencing, flow cytometry, antigen receptor repertoire analysis, and unbiased serum proteomics, which collectively identified a signature in MIS-C patients that correlated with disease severity. Despite having no evidence of active infection, MIS-C patients had elevated S100A-family alarmins and decreased antigen presentation signatures, indicative of myeloid dysfunction. MIS-C patients showed elevated expression of cytotoxicity genes in NK and CD8+ T cells and expansion of specific IgG-expressing plasmablasts. Clinically severe MIS-C patients displayed skewed memory T cell TCR repertoires and autoimmunity characterized by endothelium-reactive IgG. The alarmin, cytotoxicity, TCR repertoire, and plasmablast signatures we defined have potential for application in the clinic to better diagnose and potentially predict disease severity early in the course of MIS-C.


Subject(s)
COVID-19/immunology , COVID-19/pathology , SARS-CoV-2/immunology , Systemic Inflammatory Response Syndrome/immunology , Systemic Inflammatory Response Syndrome/pathology , Adolescent , Alarmins/immunology , Autoantibodies/immunology , CD8-Positive T-Lymphocytes/immunology , Child , Child, Preschool , Cytotoxicity, Immunologic/genetics , Endothelium/immunology , Endothelium/pathology , Humans , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Plasma Cells/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Severity of Illness Index
20.
Cell ; 162(1): 160-9, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26140596

ABSTRACT

Protective vaccines elicit high-affinity, neutralizing antibodies by selection of somatically hypermutated B cell antigen receptors (BCR) on immune complexes (ICs). This implicates Fc-Fc receptor (FcR) interactions in affinity maturation, which, in turn, are determined by IgG subclass and Fc glycan composition within ICs. Trivalent influenza virus vaccination elicited regulation of anti-hemagglutinin (HA) IgG subclass and Fc glycans, with abundance of sialylated Fc glycans (sFc) predicting quality of vaccine response. We show that sFcs drive BCR affinity selection by binding the Type-II FcR CD23, thus upregulating the inhibitory FcγRIIB on activated B cells. This elevates the threshold requirement for BCR signaling, resulting in B cell selection for higher affinity BCR. Immunization with sFc HA ICs elicited protective, high-affinity IgGs against the conserved stalk of the HA. These results reveal a novel, endogenous pathway for affinity maturation that can be exploited for eliciting high-affinity, broadly neutralizing antibodies through immunization with sialylated immune complexes.


Subject(s)
Antibodies, Neutralizing/immunology , Influenza Vaccines/immunology , Receptors, Antigen, B-Cell/immunology , Antigen-Antibody Complex/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunoglobulin Fc Fragments , Immunoglobulin G/immunology , Plasma Cells/immunology , Receptors, Antigen, B-Cell/chemistry , Receptors, Fc/metabolism , Sialic Acids/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL