Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.524
Filter
1.
Nat Immunol ; 25(8): 1422-1431, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38961274

ABSTRACT

The differentiation of naive and memory B cells into antibody-secreting cells (ASCs) is a key feature of adaptive immunity. The requirement for phosphoinositide 3-kinase-delta (PI3Kδ) to support B cell biology has been investigated intensively; however, specific functions of the related phosphoinositide 3-kinase-gamma (PI3Kγ) complex in B lineage cells have not. In the present study, we report that PI3Kγ promotes robust antibody responses induced by T cell-dependent antigens. The inborn error of immunity caused by human deficiency in PI3Kγ results in broad humoral defects, prompting our investigation of roles for this kinase in antibody responses. Using mouse immunization models, we found that PI3Kγ functions cell intrinsically within activated B cells in a kinase activity-dependent manner to transduce signals required for the transcriptional program supporting differentiation of ASCs. Furthermore, ASC fate choice coincides with upregulation of PIK3CG expression and is impaired in the context of PI3Kγ disruption in naive B cells on in vitro CD40-/cytokine-driven activation, in memory B cells on toll-like receptor activation, or in human tonsillar organoids. Taken together, our study uncovers a fundamental role for PI3Kγ in supporting humoral immunity by integrating signals instructing commitment to the ASC fate.


Subject(s)
Antibody Formation , B-Lymphocytes , Cell Differentiation , Class Ib Phosphatidylinositol 3-Kinase , Animals , Class Ib Phosphatidylinositol 3-Kinase/metabolism , Class Ib Phosphatidylinositol 3-Kinase/immunology , Mice , Cell Differentiation/immunology , Humans , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Antibody Formation/immunology , Mice, Knockout , Antibody-Producing Cells/immunology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Signal Transduction/immunology , Memory B Cells/immunology , Memory B Cells/metabolism
2.
Immunity ; 57(9): 2191-2201.e5, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39168129

ABSTRACT

Memory B cells (MBCs) formed over the individual's lifetime constitute nearly half of the circulating B cell repertoire in humans. These pre-existing MBCs dominate recall responses to their cognate antigens, but how they respond to recognition of novel antigens is not well understood. Here, we tracked the origin and followed the differentiation paths of MBCs in the early anti-spike (S) response to mRNA vaccination in SARS-CoV-2-naive individuals on single-cell and monoclonal antibody levels. Pre-existing, highly mutated MBCs showed no signs of germinal center re-entry and rapidly developed into mature antibody-secreting cells (ASCs). By contrast, and despite similar levels of S reactivity, naive B cells showed strong signs of antibody affinity maturation before differentiating into MBCs and ASCs. Thus, pre-existing human MBCs differentiate into ASCs in response to novel antigens, but the quality of the humoral and cellular anti-S response improved through the clonal selection and affinity maturation of naive precursors.


Subject(s)
Antibodies, Viral , Antibody-Producing Cells , COVID-19 Vaccines , COVID-19 , Memory B Cells , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , Memory B Cells/immunology , COVID-19/immunology , COVID-19/prevention & control , Antibodies, Viral/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibody-Producing Cells/immunology , COVID-19 Vaccines/immunology , Vaccination , Antibody Affinity/immunology , Cell Differentiation/immunology , Germinal Center/immunology , Immunologic Memory/immunology , Antibodies, Monoclonal/immunology , Adult , Female
3.
Immunity ; 56(7): 1596-1612.e4, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37164016

ABSTRACT

Antibodies produced by antibody-secreting plasma cells (ASCs) underlie multiple forms of long-lasting immunity. Here we examined the mechanisms regulating ASC turnover and persistence using a genetic reporter to time-stamp ASCs. This approach revealed ASC lifespans as heterogeneous and falling on a continuum, with only a small fraction surviving for >60 days. ASC longevity past 60 days was independent of isotype but correlated with a phenotype that developed progressively and ultimately associated with an underlying "long-lived" ASC (LL ASC)-enriched transcriptional program. While some of the differences between LL ASCs and other ASCs appeared to be acquired with age, other features were shared with some younger ASCs, such as high CD138 and CD93. Turnover was unaffected by altered ASC production, arguing against competition for niches as a major driver of turnover. Thus, ASC turnover is set by intrinsic lifespan limits, with steady-state population dynamics governed by niche vacancy rather than displacement.


Subject(s)
Longevity , Plasma Cells , Antibody-Producing Cells
4.
Immunity ; 50(3): 616-628.e6, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30850343

ABSTRACT

Humoral immunity depends on efficient activation of B cells and their subsequent differentiation into antibody-secreting cells (ASCs). The transcription factor NFκB cRel is critical for B cell proliferation, but incorporating its known regulatory interactions into a mathematical model of the ASC differentiation circuit prevented ASC generation in simulations. Indeed, experimental ectopic cRel expression blocked ASC differentiation by inhibiting the transcription factor Blimp1, and in wild-type (WT) cells cRel was dynamically repressed during ASC differentiation by Blimp1 binding the Rel locus. Including this bi-stable circuit of mutual cRel-Blimp1 antagonism into a multi-scale model revealed that dynamic repression of cRel controls the switch from B cell proliferation to ASC generation phases and hence the respective cell population dynamics. Our studies provide a mechanistic explanation of how dysregulation of this bi-stable circuit might result in pathologic B cell population phenotypes and thus offer new avenues for diagnostic stratification and treatment.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation/physiology , NF-kappa B/immunology , Animals , Antibody-Producing Cells/immunology , Cell Line , Female , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Immunity, Humoral/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
5.
Immunity ; 50(5): 1172-1187.e7, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31076359

ABSTRACT

Although viral infections elicit robust interferon-γ (IFN-γ) and long-lived antibody-secreting cell (ASC) responses, the roles for IFN-γ and IFN-γ-induced transcription factors (TFs) in ASC development are unclear. We showed that B cell intrinsic expression of IFN-γR and the IFN-γ-induced TF T-bet were required for T-helper 1 cell-induced differentiation of B cells into ASCs. IFN-γR signaling induced Blimp1 expression in B cells but also initiated an inflammatory gene program that, if not restrained, prevented ASC formation. T-bet did not affect Blimp1 upregulation in IFN-γ-activated B cells but instead regulated chromatin accessibility within the Ifng and Ifngr2 loci and repressed the IFN-γ-induced inflammatory gene program. Consistent with this, B cell intrinsic T-bet was required for formation of long-lived ASCs and secondary ASCs following viral, but not nematode, infection. Therefore, T-bet facilitates differentiation of IFN-γ-activated inflammatory effector B cells into ASCs in the setting of IFN-γ-, but not IL-4-, induced inflammatory responses.


Subject(s)
B-Lymphocytes/immunology , Interferon-gamma/immunology , Receptors, Interferon/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibody-Producing Cells/immunology , B-Lymphocytes/cytology , Cell Differentiation/immunology , Cells, Cultured , Chromatin/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Positive Regulatory Domain I-Binding Factor 1/biosynthesis , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Box Domain Proteins/genetics , Interferon gamma Receptor
6.
Nat Immunol ; 16(7): 755-65, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26006014

ABSTRACT

Acute systemic lupus erythematosus (SLE) courses with surges of antibody-secreting cells (ASCs) whose origin, diversity and contribution to serum autoantibodies remain unknown. Here, deep sequencing, proteomic profiling of autoantibodies and single-cell analysis demonstrated highly diversified ASCs punctuated by clones expressing the variable heavy-chain region VH4-34 that produced dominant serum autoantibodies. A fraction of ASC clones contained autoantibodies without mutation, a finding consistent with differentiation outside the germinal centers. A substantial ASC segment was derived from a distinct subset of newly activated naive cells of considerable clonality that persisted in the circulation for several months. Thus, selection of SLE autoreactivities occurred during polyclonal activation, with prolonged recruitment of recently activated naive B cells. Our findings shed light on the pathogenesis of SLE, help explain the benefit of agents that target B cells and should facilitate the design of future therapies.


Subject(s)
Antibody Diversity/immunology , Antibody-Producing Cells/immunology , Autoantibodies/immunology , Cell Proliferation , Lupus Erythematosus, Systemic/immunology , Acute Disease , Amino Acid Sequence , Antibody Diversity/genetics , Antibody-Producing Cells/metabolism , Autoantibodies/genetics , Autoantibodies/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , Clone Cells/immunology , Clone Cells/metabolism , Flow Cytometry , Humans , Immunoglobulin G/blood , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Immunoglobulin Variable Region/metabolism , Influenza Vaccines/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Molecular Sequence Data , Proteome/analysis , Proteome/immunology , Proteomics/methods , Sequence Homology, Amino Acid , Single-Cell Analysis/methods , Tandem Mass Spectrometry , Tetanus Toxoid/immunology
7.
Proc Natl Acad Sci U S A ; 121(29): e2404309121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38990948

ABSTRACT

Antibody-producing plasma cells fuel humoral immune responses. They also contribute to autoimmune diseases such as systemic lupus erythematosus or IgA nephropathy. Interleukin-6 and the tumor necrosis factor (TNF) family ligands BAFF (B cell-activating factor) and APRIL (a proliferation-inducing ligand) participate in plasma cell survival. BAFF binds to three receptors, BAFFR (BAFF receptor), TACI (transmembrane activator and CAML interactor), and BCMA (B cell maturation antigen), while APRIL binds to TACI, BCMA, and proteoglycans. However, which ligand-receptor pair(s) are required to maintain plasma cells in different body locations remains unknown. Here, by combining mouse genetic and pharmacological approaches, we found that plasma cells required BCMA and/or TACI but not BAFFR. BCMA responded exclusively to APRIL, while TACI responded to both BAFF and APRIL, identifying three self-sufficient ligand-receptor pairs for plasma cell maintenance: BAFF-TACI, APRIL-TACI, and APRIL-BCMA. Together, these actors accounted for 90% of circulating antibodies. In BAFF-ko mice, the reduction of plasma cells upon APRIL inhibition indicated that APRIL could function in the absence of BAFF-APRIL heteromers. No evidence was found that in the absence of BCMA and TACI, binding of APRIL to proteoglycans would help maintain plasma cells. IL-6, alone or together with BAFF and APRIL, supported mainly splenic plasmablasts and plasma cells and contributed to circulating IgG but not IgA levels. In conclusion, survival factors for plasma cells can vary with body location and with the antibody isotype that plasma cells produce. To efficiently target plasma cells, in particular IgA-producing ones, dual inhibition of BAFF and APRIL is required.


Subject(s)
B-Cell Activating Factor , B-Cell Activation Factor Receptor , B-Cell Maturation Antigen , Interleukin-6 , Transmembrane Activator and CAML Interactor Protein , Tumor Necrosis Factor Ligand Superfamily Member 13 , Animals , B-Cell Activating Factor/immunology , B-Cell Activating Factor/metabolism , B-Cell Activating Factor/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , B-Cell Maturation Antigen/immunology , B-Cell Maturation Antigen/metabolism , Transmembrane Activator and CAML Interactor Protein/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/immunology , Interleukin-6/metabolism , Interleukin-6/immunology , Mice , B-Cell Activation Factor Receptor/metabolism , B-Cell Activation Factor Receptor/immunology , B-Cell Activation Factor Receptor/genetics , Plasma Cells/immunology , Plasma Cells/metabolism , Mice, Knockout , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Mice, Inbred C57BL
8.
Nat Immunol ; 15(7): 631-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24880458

ABSTRACT

Memory B cells (MBCs) are long-lived sources of rapid, isotype-switched secondary antibody-forming cell (AFC) responses. Whether MBCs homogeneously retain the ability to self-renew and terminally differentiate or if these functions are compartmentalized into MBC subsets has remained unclear. It has been suggested that antibody isotype controls MBC differentiation upon restimulation. Here we demonstrate that subcategorizing MBCs on the basis of their expression of CD80 and PD-L2, independently of isotype, identified MBC subsets with distinct functions upon rechallenge. CD80(+)PD-L2(+) MBCs differentiated rapidly into AFCs but did not generate germinal centers (GCs); conversely, CD80(-)PD-L2(-) MBCs generated few early AFCs but robustly seeded GCs. The gene-expression patterns of the subsets supported both the identity and function of these distinct MBC types. Hence, the differentiation and regeneration of MBCs are compartmentalized.


Subject(s)
B-Lymphocyte Subsets/immunology , B7-1 Antigen/physiology , Immunoglobulin Isotypes/physiology , Immunologic Memory , Programmed Cell Death 1 Ligand 2 Protein/physiology , Amino Acid Sequence , Animals , Antibody-Producing Cells/physiology , Germinal Center/immunology , Immunization , Mice , Mice, Inbred BALB C , Molecular Sequence Data , T-Lymphocytes/physiology
9.
Immunity ; 45(1): 60-73, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27396958

ABSTRACT

Durable antibody production after vaccination or infection is mediated by long-lived plasma cells (LLPCs). Pathways that specifically allow LLPCs to persist remain unknown. Through bioenergetic profiling, we found that human and mouse LLPCs could robustly engage pyruvate-dependent respiration, whereas their short-lived counterparts could not. LLPCs took up more glucose than did short-lived plasma cells (SLPCs) in vivo, and this glucose was essential for the generation of pyruvate. Glucose was primarily used to glycosylate antibodies, but glycolysis could be promoted by stimuli such as low ATP levels and the resultant pyruvate used for respiration by LLPCs. Deletion of Mpc2, which encodes an essential component of the mitochondrial pyruvate carrier, led to a progressive loss of LLPCs and of vaccine-specific antibodies in vivo. Thus, glucose uptake and mitochondrial pyruvate import prevent bioenergetic crises and allow LLPCs to persist. Immunizations that maximize these plasma cell metabolic properties might thus provide enduring antibody-mediated immunity.


Subject(s)
Antibody-Producing Cells/immunology , Glucose/metabolism , Mitochondria/metabolism , Plasma Cells/immunology , Pyruvic Acid/metabolism , Animals , Biological Transport, Active , Cell Respiration , Cells, Cultured , Glycosylation , Humans , Immunoglobulins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Proprotein Convertase 2/genetics , Proprotein Convertase 2/metabolism , Stress, Physiological/immunology
10.
J Immunol ; 210(9): 1272-1280, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36939374

ABSTRACT

Opioid use disorders (OUDs) are a public health concern in the United States and worldwide. Current medications for OUDs may trigger side effects and are often heavily regulated. A novel treatment strategy to be used alone or in combination with existing medications is active immunization with antiopioid vaccines, which stimulate production of opioid-specific Abs that bind to the target drug and prevent its distribution to the brain. Although antiopioid vaccines have shown promising preclinical efficacy, prior clinical evaluations of vaccines targeting stimulants indicate that efficacy is limited to a subset of subjects who achieve optimal Ab responses. We have previously reported that depletion of IL-4 with a mAb increased opioid-specific IgG2a and total IgG, and it increased the number of germinal centers and germinal center T follicular helper cells in response to antiopioid vaccines via type I IL-4 signaling. The current study further investigates the mechanisms associated with IL-4-mediated increases in efficacy and whether IL-4 depletion affects specific processes involved in germinal center formation, including affinity maturation, class switching, and plasma cell differentiation in mice. Additionally, results demonstrate that preimmunization production of IL-4 after ex vivo whole blood stimulation predicted in vivo vaccine-induced Ab titers in outbred mice. Such mechanistic studies are critical for rational design of next-generation vaccine formulations, and they support the use of IL-4 as a predictive biomarker in ongoing OUD vaccine clinical studies.


Subject(s)
Interleukin-4 , Vaccines , Mice , Animals , Analgesics, Opioid , Germinal Center , Antibody-Producing Cells , Cell Proliferation
11.
Mol Cell Proteomics ; 22(2): 100492, 2023 02.
Article in English | MEDLINE | ID: mdl-36623694

ABSTRACT

Single-cell technologies are currently widely applied to obtain a deeper understanding of the phenotype of single-cells in heterogenous mixtures. However, integrated multilayer approaches including simultaneous detection of mRNA, protein expression, and intracellular phospho-proteins are still challenging. Here, we combined an adapted method to in vitro-differentiate peripheral B-cells into antibody-secreting cells (ASCs) (i.e., plasmablasts and plasma cells) with integrated multi-omic single-cell sequencing technologies to detect and quantify immunoglobulin subclass-specific surface markers, transcriptional profiles, and signaling transduction pathway components. Using a common set of surface proteins, we integrated two multimodal datasets to combine mRNA, protein expression, and phospho-protein detection in one integrated dataset. Next, we tested whether ASCs that only seem to differ in its ability to secrete different IgM, IgA, or IgG antibodies exhibit other differences that characterize these different ASCs. Our approach detected differential expression of plasmablast and plasma cell markers, homing receptors, and TNF receptors. In addition, differential sensitivity was observed for the different cytokine stimulations that were applied during in vitro differentiation. For example, IgM ASCs were more sensitive to IL-15, while IgG ASC responded more to IL-6 and IFN addition. Furthermore, tonic BCR activity was detected in IgA and IgM ASCs, while IgG ASC exhibited active BCR-independent SYK activity and NF-κB and mTOR signaling. We confirmed these findings using flow cytometry and small molecules inhibitors, demonstrating the importance of SYK, NF-κB, and mTOR activity for plasmablast/plasma cell differentiation/survival and/or IgG secretion. Taken together, our integrated multi-omics approach allowed high-resolution phenotypic characterization of single cells in a heterogenous sample of in vitro-differentiated human ASCs. Our strategy is expected to further our understanding of human ASCs in healthy and diseased samples and provide a valuable tool to identify novel biomarkers and potential drug targets.


Subject(s)
Antibody-Producing Cells , Signal Transduction , Single-Cell Gene Expression Analysis , Humans , Antibody-Producing Cells/metabolism , Immunoglobulin A , Immunoglobulin G , Immunoglobulin M , NF-kappa B , Phenotype , RNA , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases
12.
J Allergy Clin Immunol ; 153(2): 527-532, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37898408

ABSTRACT

BACKGROUND: Chronic rhinosinusitis with nasal polyps (CRSwNP) causes nasal obstruction and olfactory dysfunction. Aspirin-exacerbated respiratory disease (AERD) is the triad of CRSwNP, asthma, and respiratory reactions to COX-1 inhibitors. Patients with AERD have elevated nasal IL-5 levels and high numbers of antibody-secreting cells (ASCs), including plasma cells and plasmablasts, in their polyp tissue; in addition, their nasal polyp (NP) IgE levels are correlated with disease severity and recurrence of nasal polyposis. OBJECTIVE: We sought to explore differences in the transcriptomic profile, activation markers, and IL-5Rα expression and function of NP ASCs from patients with AERD and CRSwNP. METHODS: NP tissue was collected from patients with AERD and CRSwNP and digested into single-cell suspensions. NP cells were analyzed for protein expression by mass cytometry. For IL-5Rα functional studies, plasma cells were purified and cultured in vitro with or without IL-5 and analyzed by bulk RNA sequencing. RESULTS: Compared with polyp tissue from patients with CRSwNP, polyp tissue from patients with AERD contained significantly more ASCs and had increased ASC expression of IL-5Rα. ASCs from patients with AERD expressed higher protein levels of B-cell activation and regulatory markers (CD40, CD19, CD32, and CD38) and the proliferation marker Ki-67. ASCs from patients with AERD also expressed more IL5RA, IGHE, and cell cycle- and proliferation-related transcripts (CCND2, MKI67, CDC25A, and CDC25B) than did ASCs from patients with CRSwNP. Stimulation of plasma cells from patients with AERD with IL-5 induced key cell cycle genes (CCND2 and PTP4A3), whereas IL-5 stimulation of ASCs from patients with CRSwNP induced few transcriptomic changes. CONCLUSION: NP tissue ASCs from patients with AERD express higher levels of functional IL-5Rα and markers associated with cell cycling and proliferation than do ASCs from patients with aspirin-tolerant CRSwNP.


Subject(s)
Asthma, Aspirin-Induced , Nasal Polyps , Rhinitis , Sinusitis , Humans , Nasal Polyps/metabolism , Interleukin-5 , Rhinitis/metabolism , Asthma, Aspirin-Induced/metabolism , Aspirin/adverse effects , Chronic Disease , Antibody-Producing Cells/metabolism , Sinusitis/metabolism , Cell Proliferation , Neoplasm Proteins , Protein Tyrosine Phosphatases
13.
Immunol Rev ; 303(1): 154-167, 2021 09.
Article in English | MEDLINE | ID: mdl-34351644

ABSTRACT

Central to protective humoral immunity is the activation of B cells and their terminal differentiation into antibody-secreting plasma cells. Long-lived plasma cells (LLPC) may survive for years to decades. Such long-lived plasma cells are also responsible for producing pathogenic antibodies that cause a variety of challenges such as autoimmunity, allograft rejection, and drug neutralization. Up to now, various therapeutic strategies aimed at durably eliminating pathogenic antibodies have failed, in large part due to their inability to efficiently target LLPCs. Several antibody-based therapies have recently gained regulatory approval or are in clinical phases of development for the treatment of multiple myeloma, a malignancy of plasma cells. We discuss the exciting potential of using these emerging cancer immunotherapies to solve the antibody problem.


Subject(s)
B-Lymphocytes , Plasma Cells , Antibody-Producing Cells , Autoimmunity , Immunity, Humoral
14.
Immunol Rev ; 303(1): 52-61, 2021 09.
Article in English | MEDLINE | ID: mdl-34313339

ABSTRACT

Antibody-secreting plasma cells are a central component of short- and long-term adaptive immunity. Yet, many fundamental questions about how activated B cells decide to yield functional plasma cells have yet to be answered. Likewise, the biochemical processes underpinning the ability of plasma cells to generate and secrete large numbers of antibodies, the capacity of some plasma cell to sustain antibody secretion, presumably without interruption, for decades, and the capacity of long-lived plasma cells to avoid apoptosis despite the high-energy demands associated with sustained robust antibody synthesis and secretion each remain mysterious processes. Our objective here is to review what is currently known about these processes with an emphasis on the earliest phases of plasma cell genesis. Along the way, we will work toward developing a model that ties the biochemistry of plasma cell function and survival. The chief idea imbedded in this model is that progress toward understanding plasma cell survival mechanisms may require increased focus on the unique cell autonomous processes inherent in plasma cell differentiation and function.


Subject(s)
B-Lymphocytes , Plasma Cells , Antibody Formation , Antibody-Producing Cells , Cell Differentiation , Lymphocyte Activation
15.
Immunol Rev ; 303(1): 23-34, 2021 09.
Article in English | MEDLINE | ID: mdl-34109653

ABSTRACT

Antibodies are an essential element of the immune response to infection, and in long-term protection upon re-exposure to the same micro-organism. Antibodies are produced by plasmablasts and plasma cells, the terminally differentiated cells of the B lymphocyte lineage. These relatively rare populations, collectively termed antibody secreting cells (ASCs), have developed highly specialized transcriptional and metabolic pathways to facilitate their extraordinarily high rates of antibody synthesis and secretion. In this review, we discuss the gene regulatory network that controls ASC identity and function, with a particular focus on the processes that influence the transcription, translation, folding, modification and secretion of antibodies. We will address how ASCs have adapted their transcriptional, metabolic and protein homeostasis pathways to sustain such high rates of antibody production, and the roles that the major ASC regulators, the transcription factors, Irf4, Blimp-1 and Xbp1, play in co-ordinating these processes.


Subject(s)
Gene Regulatory Networks , Plasma Cells , Antibody-Producing Cells , B-Lymphocytes , Cell Differentiation
16.
Immunol Rev ; 303(1): 138-153, 2021 09.
Article in English | MEDLINE | ID: mdl-34337772

ABSTRACT

Antibody-secreting cells (ASC) are the effectors of protective humoral immunity and the only cell type that produces antibodies or immunoglobulins in mammals. In addition to their formidable capacity to secrete massive quantities of proteins, ASC are terminally differentiated and have unique features to become long-lived plasma cells (LLPC). Upon antigen encounter, B cells are activated through a complex multistep process to undergo fundamental morphological, subcellular, and molecular transformation to become an efficient protein factory with lifelong potential. The ASC survival potential is determined by factors at the time of induction, capacity to migration from induction to survival sites, and ability to mature in the specialized bone marrow microenvironments. In the past decade, considerable progress has been made in identifying factors regulating ASC longevity. Here, we review the intrinsic drivers, trafficking signals, and extrinsic regulators with particular focus on how they impact the survival potential to become a LLPC.


Subject(s)
Antibody-Producing Cells , Plasma Cells , Animals , B-Lymphocytes , Bone Marrow , Cell Survival , Immunity, Humoral
17.
Immunol Rev ; 303(1): 8-22, 2021 09.
Article in English | MEDLINE | ID: mdl-34010461

ABSTRACT

Humoral immunity provides protection from pathogenic infection and is mediated by antibodies following the differentiation of naive B cells (nBs) to antibody-secreting cells (ASCs). This process requires substantial epigenetic and transcriptional rewiring to ultimately repress the nB program and replace it with one conducive to ASC physiology and function. Notably, these reprogramming events occur within the framework of cell division. Efforts to understand the relationship of cell division with reprogramming and ASC differentiation in vivo have uncovered the timing and scope of reprogramming, as well as key factors that influence these events. Herein, we discuss the unique physiology of ASC and how nBs undergo epigenetic and genome architectural reorganization to acquire the necessary functions to support antibody production. We also discuss the stage-wise manner in which reprogramming occurs across cell divisions and how key molecular determinants can influence B cell fate outcomes.


Subject(s)
Antibody-Producing Cells , Plasma Cells , B-Lymphocytes , Cell Differentiation/genetics , Epigenesis, Genetic , Gene Expression Regulation
18.
Immunol Rev ; 303(1): 103-118, 2021 09.
Article in English | MEDLINE | ID: mdl-34145601

ABSTRACT

Antibody-secreting cells (ASCs) are considered work horses of the humoral immune response for their tireless effort to produce large amounts of antibodies that fulfill an array of functions in host defense, inflammation, and maintenance of homeostasis. While traditionally considered largely senescent cells, surprising recent findings demonstrate that subsets of ASCs downmodulate ongoing immune responses independent of antibody formation. Such regulatory ASCs produce IL-10 or IL-35 and are implicated in maintaining tissue and immune homeostasis. They also serve to suppress pathogenic leukocytes in infection, allergy, and inflammatory diseases that affect tissues, such as the central nervous system and the respiratory tract. Additionally, regulatory ASCs infiltrate various cancer types and restrict effective anti-tumor T cell responses. While incompletely understood, there is significant overlap in factors that control ASC differentiation, IL-10 expression by B cells and the generation of ASCs that secrete both antibodies and IL-10. In this review, we will cover the biology, phenotype, generation, maintenance and function of regulatory ASCs in various tissues under pathological and steady states. An improved understanding of the development of regulatory ASCs and their biological roles will be critical for generating novel ASC-targeted therapies for the treatment of inflammatory diseases, infection, and cancer.


Subject(s)
Antibody-Producing Cells , Neoplasms , Animals , B-Lymphocytes , Horses , Immunity, Humoral , Inflammation , Neoplasms/therapy
19.
Immunology ; 173(2): 339-359, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38934051

ABSTRACT

Maintaining intracellular redox balance is essential for the survival, antibody secretion, and mucosal immune homeostasis of immunoglobulin A (IgA) antibody-secreting cells (ASCs). However, the relationship between mitochondrial metabolic enzymes and the redox balance in ASCs has yet to be comprehensively studied. Our study unveils the pivotal role of mitochondrial enzyme PCK2 in regulating ASCs' redox balance and intestinal homeostasis. We discover that PCK2 loss, whether globally or in B cells, exacerbates dextran sodium sulphate (DSS)-induced colitis due to increased IgA ASC cell death and diminished antibody production. Mechanistically, the absence of PCK2 diverts glutamine into the TCA cycle, leading to heightened TCA flux and excessive mitochondrial reactive oxygen species (mtROS) production. In addition, PCK2 loss reduces glutamine availability for glutathione (GSH) synthesis, resulting in a decrease of total glutathione level. The elevated mtROS and reduced GSH expose ASCs to overwhelming oxidative stress, culminating in cell apoptosis. Crucially, we found that the mitochondria-targeted antioxidant Mitoquinone (Mito-Q) can mitigate the detrimental effects of PCK2 deficiency in IgA ASCs, thereby alleviating colitis in mice. Our findings highlight PCK2 as a key player in IgA ASC survival and provide a potential new target for colitis treatment.


Subject(s)
Colitis , Homeostasis , Mitochondria , Oxidative Stress , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/immunology , Mice , Mitochondria/metabolism , Immunoglobulin A/metabolism , Dextran Sulfate , Mice, Knockout , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Glutathione/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/immunology , Intestines/immunology , Apoptosis , Disease Models, Animal
20.
Blood ; 140(16): 1803-1815, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36070233

ABSTRACT

Platelet transfusion and transplantation of allogeneic stem cells and solid organs are life-saving therapies. Unwanted alloantibodies to nonself human leukocyte antigens (HLAs) on donor cells increase the immunological barrier to these therapies and are important causes of platelet transfusion refractoriness and graft rejection. Although the specificities of anti-HLA antibodies can be determined at the allelic level, traditional treatments for antibody-mediated rejection nonselectively suppress humoral immunity and are not universally successful. We designed HLA-Fc fusion proteins with a bivalent targeting module derived from extracellular domains of HLA and an Fc effector module from mouse IgG2a. We found that HLA-Fc with A2 (A2Fc) and B7 (B7Fc) antigens lowered HLA-A2- and HLA-B7-specific reactivities, respectively, in sera from HLA-sensitized patients. A2Fc and B7Fc bound to B-cell hybridomas bearing surface immunoglobulins with cognate specificities and triggered antigen-specific and Fc-dependent cytotoxicity in vitro. In immunodeficient mice carrying HLA-A2-specific hybridoma cells, A2Fc treatment lowered circulating anti-HLA-A2 levels, abolished the outgrowth of hybridoma cells, and prolonged survival compared with control groups. In an in vivo anti-HLA-A2-mediated platelet transfusion refractoriness model, A2Fc treatment mitigated refractoriness. These results support HLA-Fc being a novel strategy for antigen-specific humoral suppression to improve transfusion and transplantation outcomes. With the long-term goal of targeting HLA-specific memory B cells for desensitization, further studies of HLA-Fc's efficacy in immune-competent animal models are warranted.


Subject(s)
Isoantibodies , Thrombocytopenia , Humans , Mice , Animals , HLA-B7 Antigen , HLA Antigens , Graft Rejection , Antilymphocyte Serum , HLA-A2 Antigen , Antibody-Producing Cells , Immunoglobulin G , Receptors, Antigen, B-Cell
SELECTION OF CITATIONS
SEARCH DETAIL