Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.564
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 35: 255-284, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28142324

ABSTRACT

We comprehensively review memory B cells (MBCs), covering the definition of MBCs and their identities and subsets, how MBCs are generated, where they are localized, how they are maintained, and how they are reactivated. Whereas naive B cells adopt multiple fates upon stimulation, MBCs are more restricted in their responses. Evolving work reveals that the MBC compartment in mice and humans consists of distinct subpopulations with differing effector functions. We discuss the various approaches to define subsets and subset-specific roles. A major theme is the need to both deliver faster effector function upon reexposure and readapt to antigenically variant pathogens while avoiding burnout, which would be the result if all MBCs generated only terminal effector function. We discuss cell-intrinsic differences in gene expression and signaling that underlie differences in function between MBCs and naive B cells and among MBC subsets and how this leads to memory responses.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Immunologic Memory , Vaccines/immunology , Animals , Humans , Immunity, Humoral , Lymphocyte Activation , Mice , Transcriptome
2.
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
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 ; 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
5.
Immunity ; 53(3): 548-563.e8, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32857950

ABSTRACT

How antigen valency affects B cells in vivo during immune responses is not well understood. Here, using HIV immunogens with defined valencies ranging from 1 to 60, we investigated the role of antigen valency during different phases of B cell responses in vivo. Highly multimerized immunogens preferentially rapidly activated cognate B cells, with little affinity discrimination. This led to strong early induction of the transcription factors IRF4 (interferon regulatory factor 4) and Bcl6, driving both early extrafollicular plasma cell and germinal center responses, in a CD4+ T-cell-dependent manner, involving B cells with a broad range of affinities. Low-valency antigens induced smaller effector B cell responses, with preferential recruitment of high-affinity B cells. Thus, antigen valency has multifaceted effects on B cell responses and can dictate affinity thresholds and competitive landscapes for B cells in vivo, with implications for vaccine design.


Subject(s)
Antibody Affinity/immunology , Antigens/immunology , B-Lymphocytes/immunology , Binding Sites, Antibody/immunology , Germinal Center/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation/physiology , Interferon Regulatory Factors/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Plasma Cells/immunology , Protein Multimerization/immunology , Proto-Oncogene Proteins c-bcl-6/immunology
6.
Immunity ; 51(2): 351-366.e6, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31303400

ABSTRACT

Aging results in increased myelopoiesis, which is linked to the increased incidence of myeloid leukemias and production of myeloid-derived suppressor cells. Here, we examined the contribution of plasma cells (PCs) to age-related increases in myelopoiesis, as PCs exhibit immune regulatory function and sequester in bone marrow (BM). PC number was increased in old BM, and they exhibited high expression of genes encoding inflammatory cytokines and pathogen sensors. Antibody-mediated depletion of PCs from old mice reduced the number of myeloid-biased hematopoietic stem cells and mature myeloid cells to levels in young animals, but lymphopoiesis was not rejuvenated, indicating that redundant mechanisms inhibit that process. PCs also regulated the production of inflammatory factors from BM stromal cells, and disruption of the PC-stromal cell circuitry with inhibitors of the cytokines IL-1 and TNF-α attenuated myelopoiesis in old mice. Thus, the age-related increase in myelopoiesis is driven by an inflammatory network orchestrated by PCs.


Subject(s)
Aging/physiology , Bone Marrow/physiology , Hematopoietic Stem Cells/pathology , Inflammation/metabolism , Myelopoiesis/physiology , Plasma Cells/physiology , Animals , Cells, Cultured , Humans , Interleukin-1/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Necrosis Factor-alpha/metabolism
7.
CA Cancer J Clin ; 71(2): 176-190, 2021 03.
Article in English | MEDLINE | ID: mdl-33165928

ABSTRACT

The application of genomic profiling assays using plasma circulating tumor DNA (ctDNA) is rapidly evolving in the management of patients with advanced solid tumors. Diverse plasma ctDNA technologies in both commercial and academic laboratories are in routine or emerging use. The increasing integration of such testing to inform treatment decision making by oncology clinicians has complexities and challenges but holds significant potential to substantially improve patient outcomes. In this review, the authors discuss the current role of plasma ctDNA assays in oncology care and provide an overview of ongoing research that may inform real-world clinical applications in the near future.


Subject(s)
Biomarkers, Tumor/blood , Circulating Tumor DNA/blood , Medical Oncology/methods , Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , Clinical Decision-Making , Humans , Liquid Biopsy/methods , Liquid Biopsy/standards , Liquid Biopsy/trends , Medical Oncology/standards , Medical Oncology/trends , Mutation , Neoplasm Staging/methods , Neoplasm Staging/trends , Neoplasms/blood , Neoplasms/genetics , Neoplasms/therapy , Practice Guidelines as Topic , Societies, Medical/standards , United States
8.
Immunity ; 49(1): 120-133.e9, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30005826

ABSTRACT

B lymphocytes can suppress immunity through interleukin (IL)-10 production in infectious, autoimmune, and malignant diseases. Here, we have identified a natural plasma cell subset that distinctively expresses the inhibitory receptor LAG-3 and mediates this function in vivo. These plasma cells also express the inhibitory receptors CD200, PD-L1, and PD-L2. They develop from various B cell subsets in a B cell receptor (BCR)-dependent manner independently of microbiota in naive mice. After challenge they upregulate IL-10 expression via a Toll-like receptor-driven mechanism within hours and without proliferating. This function is associated with a unique transcriptome and epigenome, including the lowest amount of DNA methylation at the Il10 locus compared to other B cell subsets. Their augmented accumulation in naive mutant mice with increased BCR signaling correlates with the inhibition of memory T cell formation and vaccine efficacy after challenge. These natural regulatory plasma cells may be of broad relevance for disease intervention.


Subject(s)
Antigens, CD/genetics , Gene Expression , Interleukin-10/biosynthesis , Plasma Cells/immunology , Animals , Antigens, CD/immunology , B-Lymphocyte Subsets/immunology , Epigenesis, Genetic , Female , Gene Expression Profiling , Interleukin-10/genetics , Lymphocyte Activation , Male , Mice , Plasma Cells/physiology , Receptors, Antigen, B-Cell/metabolism , Salmonella Infections, Animal/immunology , Signal Transduction , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism , Up-Regulation/genetics , Vaccines/immunology , Lymphocyte Activation Gene 3 Protein
9.
Semin Immunol ; 65: 101707, 2023 01.
Article in English | MEDLINE | ID: mdl-36527759

ABSTRACT

Immuno-oncology has traditionally focused on the cellular arm of the adaptive immune response, while attributing tumor-promoting activity to humoral responses in tumor-bearing hosts. This view stems from mouse models that do not necessarily recapitulate the antibody response process consistently observed in most human cancers. In recent years, the field has reconsidered the coordinated action of T and B cell responses in the context of anti-tumor immunity, as in any other immune response. Thus, recent studies in human cancer identify B cell responses with better outcome, typically in association with superior T cell responses. An area of particular interest is tertiary lymphoid structures, where germinal centers produce isotype switched antibodies and B cells and T lymphocytes interact with other immune cell types. The presence of these lymphoid structures is associated with better immunotherapeutic responses and remain poorly understood. Here, we discuss recent discoveries on how coordination between humoral and cellular responses is required for effective immune pressure against malignant progression, providing a perspective on the role of tertiary lymphoid structures and interventions to elicit their formation in unresectable tumors.


Subject(s)
B-Lymphocytes , Neoplasms , T-Lymphocytes , Tertiary Lymphoid Structures , Animals , Humans , Mice , Adaptive Immunity/immunology , B-Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Tertiary Lymphoid Structures/immunology
10.
Proc Natl Acad Sci U S A ; 121(13): e2309994121, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38517976

ABSTRACT

Maternal immunoglobulins of the class G (IgGs) protect offspring from enteric infection, but when, where, and how these antibodies are physiologically generated and confer protection remains enigmatic. We found that circulating IgGs in adult mice preferentially bind early-life gut commensal bacteria over their own adult gut commensal bacteria. IgG-secreting plasma cells specific for early-life gut bacteria appear in the intestine soon after weaning, where they remain into adulthood. Manipulating exposure to gut bacteria or plasma cell development before, but not after, weaning reduced IgG-secreting plasma cells targeting early-life gut bacteria throughout life. Further, the development of this anti-gut commensal IgG response coincides with the early-life interval in which goblet cell-associated antigen passages (GAPs) are present in the colon. Offspring of dams "perturbed" by B cell ablation or reduced bacterial exposure in early life were more susceptible to enteric pathogen challenge. In contrast to current concepts, protective maternal IgGs targeted translocating gut commensals in the offspring, not the enteric pathogen. These early-life events affecting anti-commensal IgG production have intergenerational effects for protection of the offspring.


Subject(s)
B-Lymphocytes , Bacteria , Animals , Mice , Bacteria/metabolism , Goblet Cells/metabolism , Immunoglobulin G
11.
Proc Natl Acad Sci U S A ; 120(2): e2213056120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36595686

ABSTRACT

Despite the essential role of plasma cells in health and disease, the cellular mechanisms controlling their survival and secretory capacity are still poorly understood. Here, we identified the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) Sec22b as a unique and critical regulator of plasma cell maintenance and function. In the absence of Sec22b, plasma cells were hardly detectable and serum antibody titers were dramatically reduced. Accordingly, Sec22b-deficient mice fail to mount a protective immune response. At the mechanistic level, we demonstrated that Sec22b contributes to efficient antibody secretion and is a central regulator of plasma cell maintenance through the regulation of their transcriptional identity and of the morphology of the endoplasmic reticulum and mitochondria. Altogether, our results unveil an essential and nonredundant role for Sec22b as a regulator of plasma cell fitness and of the humoral immune response.


Subject(s)
Plasma Cells , SNARE Proteins , Mice , Animals , Plasma Cells/metabolism , R-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Endoplasmic Reticulum/metabolism , Biological Transport
12.
Immunol Rev ; 309(1): 40-63, 2022 08.
Article in English | MEDLINE | ID: mdl-35801537

ABSTRACT

Infection with SARS-CoV-2, the etiology of the ongoing COVID-19 pandemic, has resulted in over 450 million cases with more than 6 million deaths worldwide, causing global disruptions since early 2020. Memory B cells and durable antibody protection from long-lived plasma cells (LLPC) are the mainstay of most effective vaccines. However, ending the pandemic has been hampered by the lack of long-lived immunity after infection or vaccination. Although immunizations offer protection from severe disease and hospitalization, breakthrough infections still occur, most likely due to new mutant viruses and the overall decline of neutralizing antibodies after 6 months. Here, we review the current knowledge of B cells, from extrafollicular to memory populations, with a focus on distinct plasma cell subsets, such as early-minted blood antibody-secreting cells and the bone marrow LLPC, and how these humoral compartments contribute to protection after SARS-CoV-2 infection and immunization.


Subject(s)
COVID-19 , Antibodies, Neutralizing , Antibodies, Viral , Humans , Immunity, Humoral , Pandemics/prevention & control , Plasma Cells , SARS-CoV-2 , Vaccination
14.
Proc Natl Acad Sci U S A ; 119(25): e2121260119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35704755

ABSTRACT

Antibodies are produced across multiple isotypes with distinct properties that coordinate initial antigen clearance and confer long-term antigen-specific immune protection. Here, we interrogate the molecular programs of isotype-specific murine plasma cells (PC) following helper T cell-dependent immunization and within established steady-state immunity. We developed a single-cell-indexed and targeted molecular strategy to dissect conserved and divergent components of the rapid effector phase of antigen-specific IgM+ versus inflammation-modulating programs dictated by type 1 IgG2a/b+ PC differentiation. During antibody affinity maturation, the germinal center (GC) cycle imparts separable programs for post-GC type 2 inhibitory IgG1+ and type 1 inflammatory IgG2a/b+ PC to direct long-term cellular function. In the steady state, two subsets of IgM+ and separate IgG2b+ PC programs clearly segregate from splenic type 3 IgA+ PC programs that emphasize mucosal barrier protection. These diverse isotype-specific molecular pathways of PC differentiation control complementary modules of antigen clearance and immune protection that could be selectively targeted for immunotherapeutic applications and vaccine design.


Subject(s)
Cell Differentiation , Germinal Center , Plasma Cells , Animals , Antigens , Immunoglobulin G/genetics , Immunoglobulin M , Mice , Plasma Cells/cytology , Single-Cell Analysis , T-Lymphocytes, Helper-Inducer
15.
Immunol Rev ; 303(1): 62-71, 2021 09.
Article in English | MEDLINE | ID: mdl-34195999

ABSTRACT

This review is focused on the cellular dynamics and genomic programming of plasma cell (PC) precursors that arise during germinal center (GC) B cell responses in secondary lymphoid organs (SLOs) and give rise to PCs in the bone marrow. Considerable progress has been made in the phenotypic characterization of circulating and bone marrow PC precursors as well as their differentiated short-lived (SLPC) and long-lived (LLPC) counterparts, in the context of model antigen and vaccine responses. Importantly, it has been possible to infer the temporal dynamics of generation of PC precursors during a GC response. However, the nature of the PC precursors at their site of generation in SLOs, and their signaling and genomic states, remain to be elucidated. Our synthesis draws upon experimental studies conducted in murine models as well as in humans, the latter complemented with cell culture manipulations of PCs and their precursors. By integration of the studies in murine and human systems, which are being accelerated by new genomic methodologies, we highlight insights and hypotheses concerning the generation of PCs. This framework can be extended and explored from both fundamental and translational standpoints.


Subject(s)
B-Lymphocytes , Plasma Cells , Animals , Cell Differentiation , Genomics , Germinal Center , Humans , Mice
16.
Immunol Rev ; 303(1): 168-186, 2021 09.
Article in English | MEDLINE | ID: mdl-34254320

ABSTRACT

Solid organ transplantation is a life-saving procedure for patients with end-stage organ disease. Over the past 70 years, tremendous progress has been made in solid organ transplantation, particularly in T-cell-targeted immunosuppression and organ allocation systems. However, humoral alloimmune responses remain a major challenge to progress. Patients with preexisting antibodies to human leukocyte antigen (HLA) are at significant disadvantages in regard to receiving a well-matched organ, moreover, those who develop anti-HLA antibodies after transplantation face a significant foreshortening of renal allograft survival. Historical therapies to desensitize patients prior to transplantation or to treat posttransplant AMR have had limited effectiveness, likely because they do not significantly reduce antibody levels, as plasma cells, the source of antibody production, remain largely unaffected. Herein, we will discuss the significance of plasma cells in transplantation, aspects of their biology as potential therapeutic targets, clinical challenges in developing strategies to target plasma cells in transplantation, and lastly, novel approaches that have potential to advance the field.


Subject(s)
Kidney Transplantation , Graft Rejection , HLA Antigens , Humans , Isoantibodies , Plasma Cells
17.
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
18.
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
19.
Immunol Rev ; 303(1): 72-82, 2021 09.
Article in English | MEDLINE | ID: mdl-34396546

ABSTRACT

Immunological memory is a composite of lasting antibody titers maintained by plasma cells in conjunction with memory T and B cells. Memory B cells are a critical reservoir for plasma cell generation in the secondary response. Identification of memory B cells requires that they be distinguished from naïve, activated, and germinal center precursors and from plasma cells. Memory B cells are heterogeneous in isotype usage, immunoglobulin mutational content, and phenotypic marker expression. Phenotypic subsets of memory B cells are defined by PD-L2, CD80, and CD73 expression in mice, by CD27 and FCRL4 expression in humans and by T-bet in both mice and humans. These subsets display marked functional heterogeneity, including the ability to rapidly differentiate into plasma cells versus seed germinal centers in the secondary response. Memory B cells are located in the spleen, blood, other lymphoid organs, and barrier tissues, and recent evidence indicates that some memory B cells may be dedicated tissue-resident populations. Open questions about memory B cell longevity, renewal and progenitor-successor relationships with plasma cells are discussed.


Subject(s)
Immunity, Humoral , Plasma Cells , Animals , B-Lymphocytes , Germinal Center , Immunologic Memory , Mice
20.
Immunol Rev ; 300(1): 54-64, 2021 03.
Article in English | MEDLINE | ID: mdl-33278036

ABSTRACT

Antibody-secreting cells (ASCs) or plasma cells secrete antibodies and form a cornerstone of humoral immunity. B cells that receive activation signals in the presence or absence of T cells initiate a differentiation program that requires epigenetic and transcriptional reprogramming in order to ultimately form ASC. Reprogramming is accomplished through the interplay of transcription factors that initiate gene expression programs and epigenetic mechanisms that maintain these programs and cell fates. An important consideration is that all of these factors are operating in the context of cell division. Recent technical advances now allow mechanistic studies to move beyond genetic studies to identify the promoters and enhancer repertoires that are regulated by epigenetic mechanisms and transcription factors in rare cell types and differentiation stages in vivo. This review will detail efforts to integrate transcriptional and epigenetic changes during B cell differentiation with cell division in vivo. What has emerged is a multiphased differentiation model that requires distinct transcription factors and epigenetic programs at each step. The identification of markers that define each phase will help facilitate the manipulation of B cell differentiation for vaccine development or to treat diseases where antibodies are a component.


Subject(s)
Cues , Plasma Cells , Cell Differentiation/genetics , Epigenesis, Genetic , Lymphocyte Activation
SELECTION OF CITATIONS
SEARCH DETAIL