Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.254
Filter
1.
Nature ; 626(8001): 1102-1107, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355795

ABSTRACT

Plasma cells produce large quantities of antibodies and so play essential roles in immune protection1. Plasma cells, including a long-lived subset, reside in the bone marrow where they depend on poorly defined microenvironment-linked survival signals1. We show that bone marrow plasma cells use the ligand-gated purinergic ion channel P2RX4 to sense extracellular ATP released by bone marrow osteoblasts through the gap-junction protein pannexin 3 (PANX3). Mutation of Panx3 or P2rx4 each caused decreased serum antibodies and selective loss of bone marrow plasma cells. Compared to their wild-type counterparts, PANX3-null osteoblasts secreted less extracellular ATP and failed to support plasma cells in vitro. The P2RX4-specific inhibitor 5-BDBD abrogated the impact of extracellular ATP on bone marrow plasma cells in vitro, depleted bone marrow plasma cells in vivo and reduced pre-induced antigen-specific serum antibody titre with little posttreatment rebound. P2RX4 blockade also reduced autoantibody titre and kidney disease in two mouse models of humoral autoimmunity. P2RX4 promotes plasma cell survival by regulating endoplasmic reticulum homeostasis, as short-term P2RX4 blockade caused accumulation of endoplasmic reticulum stress-associated regulatory proteins including ATF4 and B-lineage mutation of the pro-apoptotic ATF4 target Chop prevented bone marrow plasma cell demise on P2RX4 inhibition. Thus, generating mature protective and pathogenic plasma cells requires P2RX4 signalling controlled by PANX3-regulated extracellular ATP release from bone marrow niche cells.


Subject(s)
Adenosine Triphosphate , Bone Marrow Cells , Plasma Cells , Animals , Mice , Adenosine Triphosphate/metabolism , Autoantibodies/immunology , Autoimmunity/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Lineage , Connexins/genetics , Connexins/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Mutation , Osteoblasts/metabolism , Plasma Cells/cytology , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, Purinergic P2X4/metabolism , Signal Transduction
2.
Nature ; 617(7961): 592-598, 2023 May.
Article in English | MEDLINE | ID: mdl-37011668

ABSTRACT

The primary two-dose SARS-CoV-2 mRNA vaccine series are strongly immunogenic in humans, but the emergence of highly infectious variants necessitated additional doses and the development of vaccines aimed at the new variants1-4. SARS-CoV-2 booster immunizations in humans primarily recruit pre-existing memory B cells5-9. However, it remains unclear whether the additional doses induce germinal centre reactions whereby re-engaged B cells can further mature, and whether variant-derived vaccines can elicit responses to variant-specific epitopes. Here we show that boosting with an mRNA vaccine against the original monovalent SARS-CoV-2 mRNA vaccine or the bivalent B.1.351 and B.1.617.2 (Beta/Delta) mRNA vaccine induced robust spike-specific germinal centre B cell responses in humans. The germinal centre response persisted for at least eight weeks, leading to significantly more mutated antigen-specific bone marrow plasma cell and memory B cell compartments. Spike-binding monoclonal antibodies derived from memory B cells isolated from individuals boosted with either the original SARS-CoV-2 spike protein, bivalent Beta/Delta vaccine or a monovalent Omicron BA.1-based vaccine predominantly recognized the original SARS-CoV-2 spike protein. Nonetheless, using a more targeted sorting approach, we isolated monoclonal antibodies that recognized the BA.1 spike protein but not the original SARS-CoV-2 spike protein from individuals who received the mRNA-1273.529 booster; these antibodies were less mutated and recognized novel epitopes within the spike protein, suggesting that they originated from naive B cells. Thus, SARS-CoV-2 booster immunizations in humans induce robust germinal centre B cell responses and can generate de novo B cell responses targeting variant-specific epitopes.


Subject(s)
B-Lymphocytes , COVID-19 Vaccines , COVID-19 , Germinal Center , Immunization, Secondary , Humans , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Germinal Center/cytology , Germinal Center/immunology , Plasma Cells/cytology , Plasma Cells/immunology , Memory B Cells/cytology , Memory B Cells/immunology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology
3.
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
4.
J Virol ; 96(9): e0002622, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35404084

ABSTRACT

Humoral immunity is a major component of the adaptive immune response against viruses and other pathogens with pathogen-specific antibody acting as the first line of defense against infection. Virus-specific antibody levels are maintained by continual secretion of antibody by plasma cells residing in the bone marrow. This raises the important question of how the virus-specific plasma cell population is stably maintained and whether memory B cells are required to replenish plasma cells, balancing their loss arising from their intrinsic death rate. In this study, we examined the longevity of virus-specific antibody responses in the serum of mice following acute viral infection with three different viruses: lymphocytic choriomeningitis virus (LCMV), influenza virus, and vesicular stomatitis virus (VSV). To investigate the contribution of memory B cells to the maintenance of virus-specific antibody levels, we employed human CD20 transgenic mice, which allow for the efficient depletion of B cells with rituximab, a human CD20-specific monoclonal antibody. Mice that had resolved an acute infection with LCMV, influenza virus, or VSV were treated with rituximab starting at 2 months after infection, and the treatment was continued for up to a year postinfection. This treatment regimen with rituximab resulted in efficient depletion of B cells (>95%), with virus-specific memory B cells being undetectable. There was an early transient drop in the antibody levels after rituximab treatment followed by a plateauing of the curve with virus-specific antibody levels remaining relatively stable (half-life of 372 days) for up to a year after infection in the absence of memory B cells. The number of virus-specific plasma cells in the bone marrow were consistent with the changes seen in serum antibody levels. Overall, our data show that virus-specific plasma cells in the bone marrow are intrinsically long-lived and can maintain serum antibody titers for extended periods of time without requiring significant replenishment from memory B cells. These results provide insight into plasma cell longevity and have implications for B cell depletion regimens in cancer and autoimmune patients in the context of vaccination in general and especially for COVID-19 vaccines. IMPORTANCE Following vaccination or primary virus infection, virus-specific antibodies provide the first line of defense against reinfection. Plasma cells residing in the bone marrow constitutively secrete antibodies, are long-lived, and can thus maintain serum antibody levels over extended periods of time in the absence of antigen. Our data, in the murine model system, show that virus-specific plasma cells are intrinsically long-lived but that some reseeding by memory B cells might occur. Our findings demonstrate that, due to the longevity of plasma cells, virus-specific antibody levels remain relatively stable in the absence of memory B cells and have implications for vaccination.


Subject(s)
Antibodies, Viral , Lymphocytic Choriomeningitis , Memory B Cells , Rituximab , Animals , Antibodies, Viral/blood , Humans , Immunity, Humoral , Immunologic Memory , Lymphocytic Choriomeningitis/immunology , Memory B Cells/cytology , Mice , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Plasma Cells/cytology , Rhabdoviridae Infections/immunology , Rituximab/pharmacology
7.
J Immunol ; 208(2): 257-266, 2022 01 15.
Article in English | MEDLINE | ID: mdl-35017215

ABSTRACT

This Brief Review delves into B cell responses in the context of allergy. The primary contribution of B cells to allergy is the production of IgE, the Ab isotype that triggers immediate hypersensitivity reactions through the release of mediators from mast cells and basophils. B cells may also have protective roles in allergy, such as through the production of IgG or as regulatory B cells. In this review, I focus on the basic principles of B cell differentiation and discuss features relevant to allergic immune responses. In particular, I discuss: (1) class-switch recombination; (2) plasma cell differentiation; (3) germinal centers and affinity maturation; and (4) memory B cells and recall responses, with an emphasis on IgE, IgG1, and IgG4. I also consider how B cells may contribute to allergic responses independent of Ab production-for example, by serving as APCs.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Hypersensitivity, Immediate/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin E/immunology , B-Lymphocytes, Regulatory/immunology , Basophils/immunology , Germinal Center/immunology , Humans , Hypersensitivity, Immediate/pathology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Mast Cells/immunology , Memory B Cells/immunology , Plasma Cells/cytology , Plasma Cells/immunology
8.
J Immunol ; 208(2): 514-525, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34911772

ABSTRACT

Upon encounter with Ag, B cells undergo a sequential process of differentiation to become Ab-secreting plasma cells. Although the key drivers of differentiation have been identified, research has been limited by the lack of in vitro models recapitulating the full process for murine B cells. In this study, we describe methodology using BCR or TLR ligation to obtain plasma cells that are phenotypically mature, have exited cell cycle and express a gene signature concordant with long-lived plasma cells. Dependent on the initial stimuli, the transcriptomes also show variation including the enhanced expression of matrisome components after BCR stimulation, suggestive of unique functional properties for the resultant plasma cells. Moreover, using the new culture conditions we demonstrate that alternative promoter choice regulating the expression of the master transcription factor Blimp-1/Prdm1 can be observed; when the canonical B cell promoter for Prdm1 is deleted, differentiating B cells exhibit flexibility in the choice of promoter, dictated by the initiating stimulus, with preferential maintenance of expression following exposure to TLR ligation. Thus our system provides a readily tractable model for furthering our understanding of plasma cell biology.


Subject(s)
Cell Differentiation/immunology , Plasma Cells/cytology , Plasma Cells/immunology , Positive Regulatory Domain I-Binding Factor 1/genetics , Receptors, Antigen, B-Cell/immunology , Animals , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Transcriptome/genetics
9.
Life Sci Alliance ; 5(3)2022 03.
Article in English | MEDLINE | ID: mdl-34952892

ABSTRACT

Antibody secreting cells (ASCs) circulate after vaccination and infection and migrate to the BM where a subset known as long-lived plasma cells (LLPCs) persists and secrete antibodies for a lifetime. The mechanisms by which circulating ASCs become LLPCs are not well elucidated. Here, we show that human blood ASCs have distinct morphology, transcriptomes, and epigenetics compared with BM LLPCs. Compared with blood ASCs, BM LLPCs have decreased nucleus/cytoplasm ratio but increased endoplasmic reticulum and numbers of mitochondria. LLPCs up-regulate pro-survival genes MCL1, BCL2, and BCL-XL while simultaneously down-regulating pro-apoptotic genes HRK1, CASP3, and CASP8 Consistent with reduced gene expression, the pro-apoptotic gene loci are less accessible in LLPCs. Of the pro-survival genes, only BCL2 is concordant in gene up-regulation and loci accessibility. Using a novel in vitro human BM mimetic, we show that blood ASCs undergo similar morphological and molecular changes that resemble ex vivo BM LLPCs. Overall, our study demonstrates that early-minted blood ASCs in the BM microniche must undergo morphological, transcriptional, and epigenetic changes to mature into apoptotic-resistant LLPCs.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Genomic Imprinting , Plasma Cells/cytology , Plasma Cells/metabolism , Adolescent , Adult , Antibody Formation/genetics , Antibody Formation/immunology , Apoptosis/genetics , Biomarkers , Cell Survival , Female , Genetic Heterogeneity , Histocytochemistry , Humans , Immunophenotyping , Male , Middle Aged , Plasma Cells/immunology , Plasma Cells/ultrastructure , Time Factors , Young Adult
10.
Front Immunol ; 12: 744573, 2021.
Article in English | MEDLINE | ID: mdl-34925321

ABSTRACT

B cell affinity maturation occurs in the germinal center (GC). Light-zone (LZ) GC B cells (BGC-cells) interact with follicular dendritic cells (FDCs) and compete for the limited, sequential help from T follicular helper cells needed to escape from apoptosis and complete their differentiation. The highest-affinity LZ BGC-cells enter the cell cycle and differentiate into PCs, following a dramatic epigenetic reorganization that induces transcriptome changes in general and the expression of the PRDM1 gene in particular. Human PC precursors are characterized by the loss of IL-4/STAT6 signaling and the absence of CD23 expression. Here, we studied the fate of human LZ BGC-cells as a function of their CD23 expression. We first showed that CD23 expression was restricted to the GC LZ, where it was primarily expressed by FDCs; less than 10% of tonsil LZ BGC-cells were positive. Sorted LZ BGC-cells left in culture and stimulated upregulated CD23 expression but were unable to differentiate into PCs - in contrast to cells that did not upregulate CD23 expression. An in-depth analysis (including single-cell gene expression) showed that stimulated CD23-negative LZ BGC-cells differentiated into plasmablasts and time course of gene expression changes delineates the transcriptional program that sustains PC differentiation. In particular, we identified a B cell proliferation signature supported by a transient MYC gene expression. Overall, the CD23 marker might be of value in answering questions about the differentiation of normal BGC-cells and allowed us to propose an instructive LZ BGC-cells maturation and fate model.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Germinal Center/immunology , Lymphocyte Activation/immunology , Plasma Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Germinal Center/cytology , Humans , Plasma Cells/cytology , Receptors, IgE/metabolism , Transcription, Genetic
11.
Nat Commun ; 12(1): 5761, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599177

ABSTRACT

The thymus is a central lymphoid organ primarily responsible for the development of T cells. A small proportion of B cells, however, also reside in the thymus to assist negative selection of self-reactive T cells. Here we show that the thymus of human neonates contains a consistent contingent of CD138+ plasma cells, producing all classes and subclasses of immunoglobulins with the exception of IgD. These antibody-secreting cells are part of a larger subset of B cells that share the expression of signature genes defining mouse B1 cells, yet lack the expression of complement receptors CD21 and CD35. Data from single-cell transcriptomic, clonal correspondence and in vitro differentiation assays support the notion of intrathymic CD138+ plasma cell differentiation, alongside other B cell subsets with distinctive molecular phenotypes. Lastly, neonatal thymic plasma cells also include clones reactive to commensal and pathogenic bacteria that commonly infect children born with antibody deficiency. Thus, our findings point to the thymus as a source of innate humoral immunity in human neonates.


Subject(s)
Cell Differentiation , Plasma Cells/cytology , Thymus Gland/cytology , Adult , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Fetal Blood/cytology , Gene Expression Profiling , Humans , Immunity, Innate , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Infant, Newborn , Lymphocyte Subsets/cytology , Principal Component Analysis , RNA-Seq , Single-Cell Analysis , Somatic Hypermutation, Immunoglobulin/genetics , Transcriptome/genetics
12.
J Clin Invest ; 131(20)2021 10 15.
Article in English | MEDLINE | ID: mdl-34473651

ABSTRACT

Little is known about how cells regulate and integrate distinct biosynthetic pathways governing differentiation and cell division. For B lineage cells it is widely accepted that activated cells must complete several rounds of mitosis before yielding antibody-secreting plasma cells. However, we report that marginal zone (MZ) B cells, innate-like naive B cells known to generate plasma cells rapidly in response to blood-borne bacteria, generate functional plasma cells despite cell-cycle arrest. Further, short-term Notch2 blockade in vivo reversed division-independent differentiation potential and decreased transcript abundance for numerous mTORC1- and Myc-regulated genes. Myc loss compromised plasma cell differentiation for MZ B cells, and reciprocally induced ectopic mTORC1 signaling in follicular B cells enabled division-independent differentiation and plasma cell-affiliated gene expression. We conclude that ongoing in situ Notch2/mTORC1 signaling in MZ B cells establishes a unique cellular state that enables rapid division-independent plasma cell differentiation.


Subject(s)
B-Lymphocytes/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Plasma Cells/cytology , Receptor, Notch2/physiology , Animals , Cell Differentiation , Cells, Cultured , Memory B Cells/physiology , Mice , Mice, Inbred C57BL , Mitosis , Signal Transduction/physiology
13.
Cell Rep ; 36(10): 109655, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34496253

ABSTRACT

The evolutionary strategy of transferring maternal antibodies via milk profoundly impacts the survival, lifelong health, and wellbeing of all neonates, including a pronounced impact on human breastfeeding success and infant development. While there has been increased recognition that interorgan connectivity influences the quality of a mother's milk, potentially to personalize it for her offspring, the underlying bases for these processes are incompletely resolved. Here, we define an essential role of Peyer's patches (PPs) for the generation of plasma cells that secrete maternal immunoglobulin A (IgA) into milk. Our metagenomic analysis reveals that the presence of certain residential microorganisms in the gastrointestinal (GI) tract, such as Bacteroides acidifaciens and Prevotella buccalis, is indispensable for the programming of maternal IgA synthesis prior to lactational transfer. Our data provide important insights into how the microbiome of the maternal GI environment, specifically through PPs, can be communicated to the next generation via milk.


Subject(s)
Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Milk, Human/immunology , Plasma Cells/cytology , Animals , Humans , Immunoglobulin A/immunology , Immunoglobulin A, Secretory/immunology , Mice , Peyer's Patches/immunology
14.
BMC Cancer ; 21(1): 1011, 2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34503477

ABSTRACT

BACKGROUND: Flow cytometry plays a key role in detecting bone marrow (BM) involvement in patients with diffuse large B-cell lymphoma (DLBCL). To improve its detection sensitivity, we need to explore novel markers. In this study, we detected the expression CD54 on lymphoma cells in BM specimens from DLBCL patients and clarified its diagnostic significance in BM involvement by DLBCL. METHODS: We collected BM specimens from 76 patients with DLBCL (germinal center B-cell (GCB) = 25, non-GCB = 51) and 10 control patients without lymphoma. We detected and compared the expression of CD54 on lymphoma cells and normal mature B cells by using 10-color panels. RESULTS: Normal plasma cells expressed a higher level of CD54 as compared with hematogones (p < 0.05) and normal mature B cells (p < 0.05). Among 76 patients, 23 of them (GCB = 12, non-GCB = 11) had BM involvement. Lymphoma B cells from 12 cases (GBC = 4, non-GCB = 8) expressed a higher level of CD54 compared to normal mature B cells (p < 0.05). Additionally, lymphoma cells of the non-GCB subtype frequently expressed a higher level of CD54 in comparison to the GCB subtype (p < 0.05). And the high expression of CD54 was not related to plasmacytoid differentiation. CONCLUSION: Aberrant expression of CD54 on lymphoma cells is frequently seen in patients' BM specimens involved by DLBCL, especially in the non-GCB subtype. CD54 could be used as a new marker to gate on lymphoma cells and improve the detection sensitivity of BM involvement in patients with DLBCL.


Subject(s)
Biomarkers, Tumor/analysis , Bone Marrow/chemistry , Flow Cytometry , Intercellular Adhesion Molecule-1/analysis , Lymphoma, Large B-Cell, Diffuse/chemistry , Adult , Aged , Aged, 80 and over , B-Lymphocytes/chemistry , B-Lymphocytes/metabolism , Biomarkers, Tumor/metabolism , Biopsy , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Adhesion Molecules/analysis , Cell Adhesion Molecules/metabolism , Cell Differentiation , Female , Germinal Center/chemistry , Germinal Center/metabolism , Germinal Center/pathology , Humans , Immunophenotyping , Intercellular Adhesion Molecule-1/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Plasma Cells/cytology
15.
Cell Rep ; 36(6): 109525, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34380042

ABSTRACT

Humoral immunity relies on the efficient differentiation of memory B cells (MBCs) into antibody-secreting cells (ASCs). T helper (Th) signals upregulate B cell receptor (BCR) signaling by potentiating Src family kinases through increasing CD45 phosphatase activity (CD45 PA). In this study, we show that high CD45 PA in MBCs enhances BCR signaling and is essential for their effective ASC differentiation. Mechanistically, Th signals upregulate CD45 PA through intensifying the surface binding of a CD45 ligand, Galectin-1. CD45 PA works as a sensor of T cell help and defines high-affinity germinal center (GC) plasma cell (PC) precursors characterized by IRF4 expression in vivo. Increasing T cell help in vitro results in an incremental CD45 PA increase and enhances ASC differentiation by facilitating effective induction of the transcription factors IRF4 and BLIMP1. This study connects Th signals with BCR signaling through Galectin-1-dependent regulation of CD45 PA and provides a mechanism for efficient ASC differentiation of MBCs.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation/immunology , Immunologic Memory , Leukocyte Common Antigens/metabolism , Plasma Cells/cytology , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Antibody Formation , CD40 Ligand/metabolism , Female , Galectin 1/metabolism , Germinal Center/cytology , Humans , Lymphocyte Subsets/metabolism , Mice, Inbred BALB C , Up-Regulation
16.
Mucosal Immunol ; 14(6): 1347-1357, 2021 11.
Article in English | MEDLINE | ID: mdl-34417548

ABSTRACT

Efficient IgA transcytosis is critical for the maintenance of a homeostatic microbiota. In the canonical model, locally-secreted dimeric (d)IgA reaches the polymeric immunoglobulin receptor (pIgR) on intestinal epithelium via simple diffusion. A role for integrin αE(CD103)ß7 during transcytosis has not been described, nor its expression by intestinal B cell lineage cells. We found that αE-deficient (αE-/-) mice have a luminal IgA deficit, despite normal antibody-secreting cells (ASC) recruitment, local IgA production and increased pIgR expression. This deficit was not due to dendritic cell (DC)-derived retinoic acid (RA) nor class-switching defects, as stool from RAG-/- mice reconstituted with αE-/- B cells was also IgA deficient. Flow cytometric, ultrastructural and transcriptional profiling showed that αEß7-expressing ASC represent an undescribed subset of terminally-differentiated intestinal plasma cells (PC) that establishes direct cell to cell contact with intestinal epithelium. We propose that IgA not only reaches pIgR through diffusion, but that αEß7+ PC dock with E-cadherin-expressing intestinal epithelium to directly relay IgA for transcytosis into the intestinal lumen.


Subject(s)
Immunoglobulin A/immunology , Integrins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Transcytosis/immunology , Animals , Cell Differentiation/immunology , Gene Expression , Gene Expression Regulation , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/immunology , Integrins/deficiency , Integrins/metabolism , Intestinal Mucosa/ultrastructure , Lymphocyte Activation , Mice , Mice, Knockout , Models, Biological , Plasma Cells/cytology , Plasma Cells/ultrastructure
17.
Cells ; 10(6)2021 06 15.
Article in English | MEDLINE | ID: mdl-34203839

ABSTRACT

The bone marrow (BM) is key to protective immunological memory because it harbors a major fraction of the body's plasma cells, memory CD4+ and memory CD8+ T-cells. Despite its paramount significance for the human immune system, many aspects of how the BM enables decade-long immunity against pathogens are still poorly understood. In this review, we discuss the relationship between BM survival niches and long-lasting humoral immunity, how intrinsic and extrinsic factors define memory cell longevity and show that the BM is also capable of adopting many responsibilities of a secondary lymphoid organ. Additionally, with more and more data on the differentiation and maintenance of memory T-cells and plasma cells upon vaccination in humans being reported, we discuss what factors determine the establishment of long-lasting immunological memory in the BM and what we can learn for vaccination technologies and antigen design. Finally, using these insights, we touch on how this holistic understanding of the BM is necessary for the development of modern and efficient vaccines against the pandemic SARS-CoV-2.


Subject(s)
Adaptive Immunity/physiology , Bone Marrow/physiology , Plasma Cells/cytology , T-Lymphocytes/cytology , Vaccinology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Humans , Immunity, Cellular/physiology , Immunologic Memory/physiology , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Vaccinology/methods , Vaccinology/trends
18.
Int J Mol Sci ; 22(13)2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34209213

ABSTRACT

Eosinophils are complex granulocytes with the capacity to react upon diverse stimuli due to their numerous and variable surface receptors, which allows them to respond in very different manners. Traditionally believed to be only part of parasitic and allergic/asthmatic immune responses, as scientific studies arise, the paradigm about these cells is continuously changing, adding layers of complexity to their roles in homeostasis and disease. Developing principally in the bone marrow by the action of IL-5 and granulocyte macrophage colony-stimulating factor GM-CSF, eosinophils migrate from the blood to very different organs, performing multiple functions in tissue homeostasis as in the gastrointestinal tract, thymus, uterus, mammary glands, liver, and skeletal muscle. In organs such as the lungs and gastrointestinal tract, eosinophils are able to act as immune regulatory cells and also to perform direct actions against parasites, and bacteria, where novel mechanisms of immune defense as extracellular DNA traps are key factors. Besides, eosinophils, are of importance in an effective response against viral pathogens by their nuclease enzymatic activity and have been lately described as involved in severe acute respiratory syndrome coronavirus SARS-CoV-2 immunity. The pleiotropic role of eosinophils is sustained because eosinophils can be also detrimental to human physiology, for example, in diseases like allergies, asthma, and eosinophilic esophagitis, where exosomes can be significant pathophysiologic units. These eosinophilic pathologies, require specific treatments by eosinophils control, such as new monoclonal antibodies like mepolizumab, reslizumab, and benralizumab. In this review, we describe the roles of eosinophils as effectors and regulatory cells and their involvement in pathological disorders and treatment.


Subject(s)
Eosinophils/physiology , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Asthma/immunology , Asthma/pathology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , Eosinophilic Esophagitis/drug therapy , Eosinophilic Esophagitis/immunology , Eosinophilic Esophagitis/pathology , Eosinophils/cytology , Eosinophils/immunology , Exosomes/metabolism , Extracellular Traps/metabolism , Humans , Plasma Cells/cytology , Plasma Cells/metabolism , SARS-CoV-2/isolation & purification
19.
STAR Protoc ; 2(3): 100633, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34258594

ABSTRACT

Molecular-level understanding of plasma cell (PC) differentiation has been modeled using lipopolysaccharide (LPS) stimulation in vitro. However, this system does not involve the B-cell receptor (BCR)-a critical component of B cell biology. Here, we present a protocol for in vitro PC differentiation system dependent on BCR signaling that easily scales up for cell number-demanding applications, including protein complex purification. We describe how to set up this system and detail applications for endogenous complex purification of chromatin-associated proteins. For further details on the use and execution of this protocol, please refer to Sciammas et al. (2011) and Ochiai et al. (2018, 2020).


Subject(s)
Cell Differentiation , Chromatin/metabolism , Plasma Cells/cytology , Proteins/isolation & purification , Receptors, Antigen, B-Cell/metabolism , Animals , Chromatography, Liquid/methods , Culture Media , Mice , Mice, Transgenic , Proteins/metabolism , Receptors, Antigen, B-Cell/genetics , Tandem Mass Spectrometry/methods
20.
Front Immunol ; 12: 705240, 2021.
Article in English | MEDLINE | ID: mdl-34305944

ABSTRACT

Germinal Centres (GCs) are transient structures in secondary lymphoid organs, where affinity maturation of B cells takes place following an infection. While GCs are responsible for protective antibody responses, dysregulated GC reactions are associated with autoimmune disease and B cell lymphoma. Typically, 'normal' GCs persist for a limited period of time and eventually undergo shutdown. In this review, we focus on an important but unanswered question - what causes the natural termination of the GC reaction? In murine experiments, lack of antigen, absence or constitutive T cell help leads to premature termination of the GC reaction. Consequently, our present understanding is limited to the idea that GCs are terminated due to a decrease in antigen access or changes in the nature of T cell help. However, there is no direct evidence on which biological signals are primarily responsible for natural termination of GCs and a mechanistic understanding is clearly lacking. We discuss the present understanding of the GC shutdown, from factors impacting GC dynamics to changes in cellular interactions/dynamics during the GC lifetime. We also address potential missing links and remaining questions in GC biology, to facilitate further studies to promote a better understanding of GC shutdown in infection and immune dysregulation.


Subject(s)
B-Lymphocyte Subsets/cytology , Germinal Center/cytology , Animals , Antibodies/immunology , Antigen Presentation , Apoptosis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Division , Cell Lineage , Cytokines/physiology , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/ultrastructure , Feedback, Physiological , Gene Rearrangement, B-Lymphocyte , Germinal Center/immunology , Germinal Center/ultrastructure , Humans , Infections/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Lymphopoiesis , Macrophages/immunology , Memory B Cells/metabolism , Mice , Models, Immunological , Plasma Cells/cytology , Plasma Cells/immunology , Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL
...