Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 981
Filter
1.
Proc Natl Acad Sci U S A ; 121(39): e2320537121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39302963

ABSTRACT

To respond and adapt, cells use surface receptors to sense environmental cues. While biochemical signal processing inside the cell is studied in depth, less is known about how physical processes during cell-cell contact impact signal acquisition. New experiments found that fast-evolving immune B cells in germinal centers (GCs) apply force to acquire antigen clusters prior to internalization, suggesting adaptive benefits of physical information extraction. We present a theory of stochastic antigen transfer and show that maximizing information gain via physical extraction can explain the dramatic phenotypic transition from naive to GC B cells-attenuated receptor signaling, enhanced force usage, and decentralized contact architecture. Our model suggests that binding-lifetime measurement and physical extraction serve as complementary modes of antigen recognition, greatly extending the dynamic range of affinity discrimination when combined. This physical-information framework further predicts that the optimal size of receptor clusters decreases as affinity improves, rationalizing the use of a multifocal synaptic pattern seen in GC B cells. By linking extraction dynamics to selection fidelity via discriminatory performance, we propose that cells may physically enhance information acquisition to sustain adaptive evolution.


Subject(s)
Antigens , B-Lymphocytes , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Antigens/immunology , Antigens/metabolism , Germinal Center/immunology , Germinal Center/cytology , Animals , Signal Transduction/immunology , Humans , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Models, Immunological
2.
Nat Commun ; 15(1): 7982, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39266537

ABSTRACT

Precise regulation of B cell differentiation is essential for an effective adaptive immune response. Here, we show that B cell development in mice with B cell-specific Maf deletion is unaffected, but marginal zone B cells, germinal centre B cells, and plasmablasts are significantly more frequent in the spleen of naive Maf-deficient mice compared to wild type controls. In the context of a T cell-dependent immunization, Maf deletion causes increased proliferation of germinal centre B cells and extrafollicular plasmablasts. This is accompanied by higher production of antigen-specific IgG1 antibodies with minimal modification of early memory B cells, but a reduction in plasma cell numbers. Single-cell RNA sequencing shows upregulation of genes associated with DNA replication and cell cycle progression, confirming the role of Maf in cell proliferation. Subsequent pathway analysis reveals that Maf influences cellular metabolism, transporter activity, and mitochondrial proteins, which have been implicated in controlling the germinal centre reaction. In summary, our findings demonstrate that Maf acts intrinsically in B cells as a negative regulator of late B cell differentiation, plasmablast proliferation and germinal centre B cell formation.


Subject(s)
B-Lymphocytes , Cell Differentiation , Cell Proliferation , Germinal Center , Plasma Cells , Proto-Oncogene Proteins c-maf , Animals , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/cytology , Mice , Plasma Cells/immunology , Plasma Cells/metabolism , Plasma Cells/cytology , Cell Differentiation/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Proto-Oncogene Proteins c-maf/genetics , Mice, Knockout , Mice, Inbred C57BL , Spleen/cytology , Spleen/metabolism , Spleen/immunology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Female
3.
Nat Commun ; 15(1): 6971, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39138218

ABSTRACT

Ligation of the B cell antigen receptor (BCR) initiates humoral immunity. However, BCR signaling without appropriate co-stimulation commits B cells to death rather than to differentiation into immune effector cells. How BCR activation depletes potentially autoreactive B cells while simultaneously primes for receiving rescue and differentiation signals from cognate T lymphocytes remains unknown. Here, we use a mass spectrometry-based proteomic approach to identify cytosolic/nuclear shuttling elements and uncover transcription factor EB (TFEB) as a central BCR-controlled rheostat that drives activation-induced apoptosis, and concurrently promotes the reception of co-stimulatory rescue signals by supporting B cell migration and antigen presentation. CD40 co-stimulation prevents TFEB-driven cell death, while enhancing and prolonging TFEB's nuclear residency, which hallmarks antigenic experience also of memory B cells. In mice, TFEB shapes the transcriptional landscape of germinal center B cells. Within the germinal center, TFEB facilitates the dark zone entry of light-zone-residing centrocytes through regulation of chemokine receptors and, by balancing the expression of Bcl-2/BH3-only family members, integrates antigen-induced apoptosis with T cell-provided CD40 survival signals. Thus, TFEB reprograms antigen-primed germinal center B cells for cell fate decisions.


Subject(s)
Apoptosis , B-Lymphocytes , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , CD40 Antigens , Germinal Center , Receptors, Antigen, B-Cell , Animals , Germinal Center/immunology , Germinal Center/cytology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Mice , CD40 Antigens/metabolism , CD40 Antigens/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Mice, Inbred C57BL , Lymphocyte Activation/immunology , Cell Differentiation/immunology , Signal Transduction , Antigen Presentation/immunology
4.
Methods Mol Biol ; 2826: 79-91, 2024.
Article in English | MEDLINE | ID: mdl-39017887

ABSTRACT

CRISPR-Cas9 genome editing is a powerful tool for assessing the functional role of candidate genes. In vitro CRISPR/Cas9 screens have been used to rapidly assess the role of thousands of genes in the differentiation and function of immune populations. However, the physiological relevance of a gene is often dependent on signals received in the tissue microenvironment, such as exposure to growth factors, chemokines, cytokines, and cell contact-dependent signals, which may not be recapitulated in an in vitro setting. Additionally, in vitro approaches are not sufficient to induce the differentiation of all cell populations limiting the cell types that can be screened. This has posed a major barrier to understanding the genes regulating the differentiation of germinal center B cells. Here, we describe an approach to perform an in vivo Crispr-Cas9 screen to specifically ablate genes in activated B cells. Using this approach, we have been able to reveal novel transcriptional regulators of germinal center B cell differentiation following viral infection.


Subject(s)
B-Lymphocytes , CRISPR-Cas Systems , Cell Differentiation , Gene Editing , Animals , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Cell Differentiation/genetics , Gene Editing/methods , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/cytology , Gene Deletion , RNA, Guide, CRISPR-Cas Systems/genetics
5.
Phys Rev E ; 109(6-1): 064409, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39020898

ABSTRACT

We introduce a multiscale model for affinity maturation, which aims to capture the intraclonal, interclonal, and epitope-specific organization of the B-cell population in a germinal center. We describe the evolution of the B-cell population via a quasispecies dynamics, with species corresponding to unique B-cell receptors (BCRs), where the desired multiscale structure is reflected on the mutational connectivity of the accessible BCR space, and on the statistical properties of its fitness landscape. Within this mathematical framework, we study the competition among classes of BCRs targeting different antigen epitopes, and we construct an effective immunogenic space where epitope immunodominance relations can be universally characterized. We finally study how varying the relative composition of a mixture of antigens with variable and conserved domains allows for a parametric exploration of this space, and we identify general principles for the rational design of two-antigen cocktails.


Subject(s)
Germinal Center , Receptors, Antigen, B-Cell , Germinal Center/immunology , Germinal Center/cytology , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/immunology , Models, Immunological , Immunodominant Epitopes/immunology , B-Lymphocytes/immunology
6.
Nature ; 632(8025): 637-646, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085603

ABSTRACT

Nasal vaccination elicits a humoral immune response that provides protection from airborne pathogens1, yet the origins and specific immune niches of antigen-specific IgA-secreting cells in the upper airways are unclear2. Here we define nasal glandular acinar structures and the turbinates as immunological niches that recruit IgA-secreting plasma cells from the nasal-associated lymphoid tissues (NALTs)3. Using intact organ imaging, we demonstrate that nasal vaccination induces B cell expansion in the subepithelial dome of the NALT, followed by invasion into commensal-bacteria-driven chronic germinal centres in a T cell-dependent manner. Initiation of the germinal centre response in the NALT requires pre-expansion of antigen-specific T cells, which interact with cognate B cells in interfollicular regions. NALT ablation and blockade of PSGL-1, which mediates interactions with endothelial cell selectins, demonstrated that NALT-derived IgA-expressing B cells home to the turbinate region through the circulation, where they are positioned primarily around glandular acinar structures. CCL28 expression was increased in the turbinates in response to vaccination and promoted homing of IgA+ B cells to this site. Thus, in response to nasal vaccination, the glandular acini and turbinates provide immunological niches that host NALT-derived IgA-secreting cells. These cellular events could be manipulated in vaccine design or in the treatment of upper airway allergic responses.


Subject(s)
Immunoglobulin A , Lymphoid Tissue , Nasal Mucosa , Plasma Cells , T-Lymphocytes , Turbinates , Animals , Female , Male , Mice , Bacteria/immunology , Cell Movement , Chemokines, CC/immunology , Chemokines, CC/metabolism , Germinal Center/immunology , Germinal Center/cytology , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Lymphoid Tissue/immunology , Lymphoid Tissue/cytology , Mice, Inbred C57BL , Nasal Mucosa/cytology , Nasal Mucosa/immunology , Plasma Cells/immunology , Plasma Cells/cytology , Plasma Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Turbinates/cytology , Turbinates/immunology , Vaccination , Administration, Intranasal , Vaccines/immunology , Symbiosis
7.
Nature ; 632(8025): 630-636, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085605

ABSTRACT

The upper airway is an important site of infection, but immune memory in the human upper airway is poorly understood, with implications for COVID-19 and many other human diseases1-4. Here we demonstrate that nasal and nasopharyngeal swabs can be used to obtain insights into these challenging problems, and define distinct immune cell populations, including antigen-specific memory B cells and T cells, in two adjacent anatomical sites in the upper airway. Upper airway immune cell populations seemed stable over time in healthy adults undergoing monthly swabs for more than 1 year, and prominent tissue resident memory T (TRM) cell and B (BRM) cell populations were defined. Unexpectedly, germinal centre cells were identified consistently in many nasopharyngeal swabs. In subjects with SARS-CoV-2 breakthrough infections, local virus-specific BRM cells, plasma cells and germinal centre B cells were identified, with evidence of local priming and an enrichment of IgA+ memory B cells in upper airway compartments compared with blood. Local plasma cell populations were identified with transcriptional profiles of longevity. Local virus-specific memory CD4+ TRM cells and CD8+ TRM cells were identified, with diverse additional virus-specific T cells. Age-dependent upper airway immunological shifts were observed. These findings provide new understanding of immune memory at a principal mucosal barrier tissue in humans.


Subject(s)
Immunologic Memory , Memory B Cells , Memory T Cells , Nasal Mucosa , Nasopharynx , SARS-CoV-2 , Adult , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , COVID-19/immunology , COVID-19/virology , Germinal Center/immunology , Germinal Center/cytology , Immunoglobulin A/immunology , Immunologic Memory/immunology , Memory B Cells/immunology , Memory T Cells/immunology , Nasal Mucosa/immunology , Nasal Mucosa/virology , Nasopharynx/virology , Nasopharynx/immunology , Plasma Cells/immunology , Plasma Cells/cytology , SARS-CoV-2/immunology
8.
J Math Biol ; 89(1): 10, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847854

ABSTRACT

We propose a stochastic framework to describe the evolution of the B-cell repertoire during germinal center (GC) reactions. Our model is formulated as a multitype age-dependent branching process with time-varying immigration. The immigration process captures the mechanism by which founder B cells initiate clones by gradually seeding GC over time, while the branching process describes the temporal evolution of the composition of these clones. The model assigns a type to each cell to represent attributes of interest. Examples of attributes include the binding affinity class of the B cells, their clonal family, or the nucleotide sequence of the heavy and light chains of their receptors. The process is generally non-Markovian. We present its properties, including as t → ∞ when the process is supercritical, the most relevant case to study expansion of GC B cells. We introduce temporal alpha and beta diversity indices for multitype branching processes. We focus on the dynamics of clonal dominance, highlighting its non-stationarity, and the accumulation of somatic hypermutations in the context of sequential immunization. We evaluate the impact of the ongoing seeding of GC by founder B cells on the dynamics of the B-cell repertoire, and quantify the effect of precursor frequency and antigen availability on the timing of GC entry. An application of the model illustrates how it may help with interpretation of BCR sequencing data.


Subject(s)
B-Lymphocytes , Germinal Center , Models, Immunological , Stochastic Processes , B-Lymphocytes/immunology , Humans , Germinal Center/immunology , Germinal Center/cytology , Animals , Somatic Hypermutation, Immunoglobulin/genetics , Mathematical Concepts , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology
9.
Methods Mol Biol ; 2813: 281-293, 2024.
Article in English | MEDLINE | ID: mdl-38888784

ABSTRACT

The development of an effective humoral response to pathogens and immunogens is a multiphase biological process, which is mediated by the coordinated function of specialized immune cell types in secondary lymphoid organs and particularly in T cell and follicular areas. More specifically, within the follicular/germinal center area, the orchestrated interplay between B cells, follicular helper CD4 T cells (Tfh), and stromal cells triggers a cascade of immune reactions leading to the development of memory B cells and plasma cells able to generate effective, antigen-specific antibodies. The role of Tfh cells in this process is critical. Given the need for vaccines capable to induce antibodies of high affinity, neutralizing activity, and durability, understanding the cellular and molecular mechanisms regulating Tfh cell development is of great importance. Here, we describe novel approaches for the comprehensive understanding of these cells and possible implications for future studies in vaccine development and the understanding of the pathogenesis of relevant diseases.


Subject(s)
T Follicular Helper Cells , Humans , T Follicular Helper Cells/immunology , Germinal Center/immunology , Germinal Center/cytology , B-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Cell Differentiation/immunology
10.
Curr Opin Immunol ; 87: 102421, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38733669

ABSTRACT

Effective high-affinity, long-term humoral immunity requires T cell help provided by a subset of differentiated CD4+ T cells known as T follicular helper (Tfh) cells. Classically, Tfh cells provide contact-dependent help for the generation of germinal centers (GCs) in secondary lymphoid organs (SLOs). Recent studies have expanded the conventional definition of Tfh cells, revealing new functions, new descriptions of Tfh subsets, new factors regulating Tfh differentiation, and new roles outside of SLO GCs. Together, these data suggest that one Tfh is not equivalent to another, helping redefine our understanding of Tfh cells and their biology.


Subject(s)
Cell Differentiation , Germinal Center , T Follicular Helper Cells , Cell Differentiation/immunology , Humans , Animals , T Follicular Helper Cells/immunology , Germinal Center/immunology , Germinal Center/cytology , Immunity, Humoral , T-Lymphocytes, Helper-Inducer/immunology
11.
Int J Biochem Cell Biol ; 172: 106588, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38768890

ABSTRACT

Immune responses in early life are characterized by a failure to robustly generate long-lasting protective responses against many common pathogens or upon vaccination. This is associated with a reduced ability to generate T-cell dependent high affinity antibodies. This review highlights the differences in T-cell dependent antibody responses observed between infants and adults, in particular focussing on the alterations in immune cell function that lead to reduced T follicular helper cell-B cell crosstalk within germinal centres in early life. Understanding the distinct functional characteristics of early life humoral immunity, and how these are regulated, will be critical in guiding age-appropriate immunological interventions in the very young.


Subject(s)
Immunity, Humoral , Humans , Immunity, Humoral/immunology , Animals , B-Lymphocytes/immunology , Infant , Germinal Center/immunology , Germinal Center/cytology , Adult
12.
Nature ; 627(8003): 399-406, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448581

ABSTRACT

Immune cells rely on transient physical interactions with other immune and non-immune populations to regulate their function1. To study these 'kiss-and-run' interactions directly in vivo, we previously developed LIPSTIC (labelling immune partnerships by SorTagging intercellular contacts)2, an approach that uses enzymatic transfer of a labelled substrate between the molecular partners CD40L and CD40 to label interacting cells. Reliance on this pathway limited the use of LIPSTIC to measuring interactions between CD4+ T helper cells and antigen-presenting cells, however. Here we report the development of a universal version of LIPSTIC (uLIPSTIC), which can record physical interactions both among immune cells and between immune and non-immune populations irrespective of the receptors and ligands involved. We show that uLIPSTIC can be used, among other things, to monitor the priming of CD8+ T cells by dendritic cells, reveal the steady-state cellular partners of regulatory T cells and identify germinal centre-resident T follicular helper cells on the basis of their ability to interact cognately with germinal centre B cells. By coupling uLIPSTIC with single-cell transcriptomics, we build a catalogue of the immune populations that physically interact with intestinal epithelial cells at the steady state and profile the evolution of the interactome of lymphocytic choriomeningitis virus-specific CD8+ T cells in multiple organs following systemic infection. Thus, uLIPSTIC provides a broadly useful technology for measuring and understanding cell-cell interactions across multiple biological systems.


Subject(s)
B-Lymphocytes , CD8-Positive T-Lymphocytes , Cell Communication , Dendritic Cells , Epithelial Cells , T Follicular Helper Cells , T-Lymphocytes, Regulatory , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Ligands , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , T Follicular Helper Cells/cytology , T Follicular Helper Cells/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Germinal Center/cytology , Single-Cell Gene Expression Analysis , Epithelial Cells/cytology , Epithelial Cells/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Organ Specificity
13.
Nature ; 628(8008): 612-619, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509366

ABSTRACT

There is increasing interest in how immune cells in the meninges-the membranes that surround the brain and spinal cord-contribute to homeostasis and disease in the central nervous system1,2. The outer layer of the meninges, the dura mater, has recently been described to contain both innate and adaptive immune cells, and functions as a site for B cell development3-6. Here we identify organized lymphoid structures that protect fenestrated vasculature in the dura mater. The most elaborate of these dural-associated lymphoid tissues (DALT) surrounded the rostral-rhinal confluence of the sinuses and included lymphatic vessels. We termed this structure, which interfaces with the skull bone marrow and a comparable venous plexus at the skull base, the rostral-rhinal venolymphatic hub. Immune aggregates were present in DALT during homeostasis and expanded with age or after challenge with systemic or nasal antigens. DALT contain germinal centre B cells and support the generation of somatically mutated, antibody-producing cells in response to a nasal pathogen challenge. Inhibition of lymphocyte entry into the rostral-rhinal hub at the time of nasal viral challenge abrogated the generation of germinal centre B cells and class-switched plasma cells, as did perturbation of B-T cell interactions. These data demonstrate a lymphoid structure around vasculature in the dura mater that can sample antigens and rapidly support humoral immune responses after local pathogen challenge.


Subject(s)
Dura Mater , Immunity, Humoral , Lymphoid Tissue , Veins , Administration, Intranasal , Antigens/administration & dosage , Antigens/immunology , Bone Marrow/immunology , Central Nervous System/blood supply , Central Nervous System/immunology , Dura Mater/blood supply , Dura Mater/immunology , Germinal Center/cytology , Germinal Center/immunology , Lymphatic Vessels/immunology , Lymphoid Tissue/blood supply , Lymphoid Tissue/immunology , Plasma Cells/immunology , Skull/blood supply , T-Lymphocytes/immunology , Veins/physiology , Humans , Male , Female , Adult , Middle Aged , Animals , Mice , Aged, 80 and over
14.
Nature ; 627(8003): 407-415, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38383779

ABSTRACT

Neuromyelitis optica is a paradigmatic autoimmune disease of the central nervous system, in which the water-channel protein AQP4 is the target antigen1. The immunopathology in neuromyelitis optica is largely driven by autoantibodies to AQP42. However, the T cell response that is required for the generation of these anti-AQP4 antibodies is not well understood. Here we show that B cells endogenously express AQP4 in response to activation with anti-CD40 and IL-21 and are able to present their endogenous AQP4 to T cells with an AQP4-specific T cell receptor (TCR). A population of thymic B cells emulates a CD40-stimulated B cell transcriptome, including AQP4 (in mice and humans), and efficiently purges the thymic TCR repertoire of AQP4-reactive clones. Genetic ablation of Aqp4 in B cells rescues AQP4-specific TCRs despite sufficient expression of AQP4 in medullary thymic epithelial cells, and B-cell-conditional AQP4-deficient mice are fully competent to raise AQP4-specific antibodies in productive germinal-centre responses. Thus, the negative selection of AQP4-specific thymocytes is dependent on the expression and presentation of AQP4 by thymic B cells. As AQP4 is expressed in B cells in a CD40-dependent (but not AIRE-dependent) manner, we propose that thymic B cells might tolerize against a group of germinal-centre-associated antigens, including disease-relevant autoantigens such as AQP4.


Subject(s)
Aquaporin 4 , Autoantibodies , Autoantigens , B-Lymphocytes , Immune Tolerance , Neuromyelitis Optica , Animals , Humans , Mice , AIRE Protein , Aquaporin 4/deficiency , Aquaporin 4/genetics , Aquaporin 4/immunology , Aquaporin 4/metabolism , Autoantibodies/immunology , Autoantigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/immunology , Germinal Center/cytology , Germinal Center/immunology , Neuromyelitis Optica/immunology , Neuromyelitis Optica/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Thyroid Epithelial Cells/immunology , Thyroid Epithelial Cells/metabolism , Transcriptome
15.
Nat Rev Immunol ; 24(7): 461-470, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38332373

ABSTRACT

Plasma cells are unique immune effectors, capable of producing large amounts of high-affinity antibodies that protect against pathogenic infections. Although most plasma cells have short lifespans, certain conditions or vaccinations can give rise to long-lived plasma cells (LLPCs) that provide individuals with lifelong protection against pathogen exposure. The nature of these LLPCs is poorly understood; however, recent studies have shed new light on the ontogeny, diversity, maturation and survival of these unique cells. Whereas LLPCs had been thought to arise preferentially from germinal centres, novel genetic tools have revealed that they can originate from various stages throughout the humoral response. Furthermore, new single-cell analyses have shown that mouse and human plasma cells are heterogeneous and may undergo further maturation in situ in the bone marrow niche. Finally, plasma cells were previously considered to be sessile cells maintained in fixed survival niches, but new data show that plasma cell subsets can differentially migrate and organize into clusters that may be associated with survival niches. These descriptive findings provide new insights into how cell-intrinsic programmes and extrinsic factors may regulate the longevity of plasma cells in various contexts, which suggest new research avenues for their functional validation.


Subject(s)
Cell Differentiation , Cell Survival , Plasma Cells , Plasma Cells/immunology , Plasma Cells/cytology , Humans , Animals , Cell Survival/immunology , Cell Differentiation/immunology , Mice , Germinal Center/immunology , Germinal Center/cytology
16.
J Virol ; 97(12): e0100823, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37962378

ABSTRACT

IMPORTANCE: The human gammaherpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus are etiologic agents of numerous B cell lymphomas. A hallmark of gammaherpesvirus infection is their ability to establish lifelong latency in B cells. However, the specific mechanisms that mediate chronic infection in B cells in vivo remain elusive. Cellular E3 ubiquitin ligases regulate numerous biological processes by catalyzing ubiquitylation and modifying protein location, function, or half-life. Many viruses hijack host ubiquitin ligases to evade antiviral host defense and promote viral fitness. Here, we used the murine gammaherpesvirus 68 in vivo system to demonstrate that the E3 ligase Cul4b is essential for this virus to establish latency in germinal center B cells. These findings highlight an essential role for this E3 ligase in promoting chronic gammaherpesvirus infection in vivo and suggest that targeted inhibition of E3 ligases may provide a novel and effective intervention strategy against gammaherpesvirus-associated diseases.


Subject(s)
B-Lymphocytes , Gammaherpesvirinae , Herpesviridae Infections , Persistent Infection , Animals , Mice , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cullin Proteins/metabolism , Gammaherpesvirinae/physiology , Germinal Center/cytology , Germinal Center/virology , Herpesviridae Infections/enzymology , Herpesviridae Infections/virology , Persistent Infection/enzymology , Persistent Infection/virology , Ubiquitins/metabolism , Virus Latency
17.
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
18.
Nature ; 613(7945): 735-742, 2023 01.
Article in English | MEDLINE | ID: mdl-36473496

ABSTRACT

Feedback inhibition of humoral immunity by antibodies was first documented in 19091. Subsequent studies showed that, depending on the context, antibodies can enhance or inhibit immune responses2,3. However, little is known about how pre-existing antibodies influence the development of memory B cells. Here we examined the memory B cell response in individuals who received two high-affinity anti-SARS-CoV-2 monoclonal antibodies and subsequently two doses of an mRNA vaccine4-8. We found that the recipients of the monoclonal antibodies produced antigen-binding and neutralizing titres that were only fractionally lower compared than in control individuals. However, the memory B cells of the individuals who received the monoclonal antibodies differed from those of control individuals in that they predominantly expressed low-affinity IgM antibodies that carried small numbers of somatic mutations and showed altered receptor binding domain (RBD) target specificity, consistent with epitope masking. Moreover, only 1 out of 77 anti-RBD memory antibodies tested neutralized the virus. The mechanism underlying these findings was examined in experiments in mice that showed that germinal centres formed in the presence of the same antibodies were dominated by low-affinity B cells. Our results indicate that pre-existing high-affinity antibodies bias germinal centre and memory B cell selection through two distinct mechanisms: (1) by lowering the activation threshold for B cells, thereby permitting abundant lower-affinity clones to participate in the immune response; and (2) through direct masking of their cognate epitopes. This may in part explain the shifting target profile of memory antibodies elicited by booster vaccinations9.


Subject(s)
Antibodies, Viral , B-Lymphocytes , COVID-19 Vaccines , COVID-19 , Feedback, Physiological , Immunologic Memory , Vaccination , mRNA Vaccines , Animals , Mice , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19/therapy , COVID-19/virology , SARS-CoV-2/immunology , mRNA Vaccines/immunology , COVID-19 Vaccines/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Immunoglobulin M/immunology , Germinal Center/cytology , Germinal Center/immunology , Immunization, Secondary , Somatic Hypermutation, Immunoglobulin
19.
Cell ; 186(1): 131-146.e13, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36565697

ABSTRACT

Germinal centers (GCs) form in secondary lymphoid organs in response to infection and immunization and are the source of affinity-matured B cells. The duration of GC reactions spans a wide range, and long-lasting GCs (LLGCs) are potentially a source of highly mutated B cells. We show that rather than consisting of continuously evolving B cell clones, LLGCs elicited by influenza virus or SARS-CoV-2 infection in mice are sustained by progressive replacement of founder clones by naive-derived invader B cells that do not detectably bind viral antigens. Rare founder clones that resist replacement for long periods are enriched in clones with heavily mutated immunoglobulins, including some with very high affinity for antigen, that can be recalled by boosting. Our findings reveal underappreciated aspects of the biology of LLGCs generated by respiratory virus infection and identify clonal replacement as a potential constraint on the development of highly mutated antibodies within these structures.


Subject(s)
B-Lymphocytes , Germinal Center , RNA Virus Infections , Animals , Mice , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Clone Cells , COVID-19 , Germinal Center/cytology , Germinal Center/immunology , SARS-CoV-2 , Influenza, Human , RNA Virus Infections/immunology , RNA Virus Infections/pathology , RNA Virus Infections/virology
20.
Nature ; 609(7929): 998-1004, 2022 09.
Article in English | MEDLINE | ID: mdl-36131022

ABSTRACT

Germinal centres are the engines of antibody evolution. Here, using human immunodeficiency virus (HIV) Env protein immunogen priming in rhesus monkeys followed by a long period without further immunization, we demonstrate germinal centre B (BGC) cells that last for at least 6 months. A 186-fold increase in BGC cells was present by week 10 compared with conventional immunization. Single-cell transcriptional profiling showed that both light- and dark-zone germinal centre states were sustained. Antibody somatic hypermutation of BGC cells continued to accumulate throughout the 29-week priming period, with evidence of selective pressure. Env-binding BGC cells were still 49-fold above baseline at 29 weeks, which suggests that they could remain active for even longer periods of time. High titres of HIV-neutralizing antibodies were generated after a single booster immunization. Fully glycosylated HIV trimer protein is a complex antigen, posing considerable immunodominance challenges for B cells1,2. Memory B cells generated under these long priming conditions had higher levels of antibody somatic hypermutation, and both memory B cells and antibodies were more likely to recognize non-immunodominant epitopes. Numerous BGC cell lineage phylogenies spanning more than the 6-month germinal centre period were identified, demonstrating continuous germinal centre activity and selection for at least 191 days with no further antigen exposure. A long-prime, slow-delivery (12 days) immunization approach holds promise for difficult vaccine targets and suggests that patience can have great value for tuning of germinal centres to maximize antibody responses.


Subject(s)
Antibody Affinity , B-Lymphocytes , Cell Movement , Clone Cells , Germinal Center , HIV Antibodies , Immunization , Animals , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibody Affinity/genetics , Antibody Affinity/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Clone Cells/cytology , Clone Cells/immunology , Epitopes, B-Lymphocyte/immunology , Gene Expression Profiling , Germinal Center/cytology , Germinal Center/immunology , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , Humans , Immunization, Secondary , Macaca mulatta/immunology , Macaca mulatta/virology , Memory B Cells/cytology , Memory B Cells/immunology , Single-Cell Analysis , Somatic Hypermutation, Immunoglobulin/genetics , Somatic Hypermutation, Immunoglobulin/immunology , Time Factors , env Gene Products, Human Immunodeficiency Virus/administration & dosage , env Gene Products, Human Immunodeficiency Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL