Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.587
Filter
1.
J Exp Med ; 221(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-39093312

ABSTRACT

Adaptive immune cell function is regulated by a highly diverse receptor recombined from variable germline-encoded segments that can recognize an almost unlimited array of epitopes. While this diversity enables the recognition of any pathogen, it also poses a risk of self-recognition, leading to autoimmunity. Many layers of regulation are present during both the generation and activation of B cells to prevent this phenomenon, although they are evidently imperfect. In recent years, our ability to analyze immune repertoires at scale has drastically increased, both through advances in sequencing and single-cell analyses. Here, we review the current knowledge on B cell repertoire analyses, focusing on their implication for autoimmunity. These studies demonstrate that a failure of tolerance occurs at multiple independent checkpoints in different autoimmune contexts, particularly during B cell maturation, plasmablast differentiation, and within germinal centers. These failures are marked by distinct repertoire features that may be used to identify disease- or patient-specific therapeutic approaches.


Subject(s)
Autoimmunity , B-Lymphocytes , Immune Tolerance , Humans , Autoimmunity/immunology , B-Lymphocytes/immunology , Animals , Immune Tolerance/immunology , Germinal Center/immunology , Cell Differentiation/immunology
2.
Article in Chinese | MEDLINE | ID: mdl-39118513

ABSTRACT

Objective:To investigate the clinical features, imaging findings, pathological phenotype, treatment and prognosis of unicentric Castleman disease in the children's neck, in order to improve the understanding of CD among Otolaryngology Head and Neck Surgery. Methods:Retrospective cross-sectional, observational study was undertaken in Kunming Children's Hospital, from the archival data between January July 2015 and June 2020. Only 6 cases of CD were identified after studying the histomorphological characteristics and neck mass diagnosed. The imaging and pathological features were summarized and the pathogenesis was discussed. Results:Among the 6 cases of Castleman disease, five were male and one was female. Histopathology: Five cases were hyaline vascular subtype, one was mixed type. The uniform clinicopathologic features seen in all hyaline vascular subtype of CD included atrophic germinal centre with lymphocyte depletion, concentric rings of small lymphocytes, increased vascularity and predominance of high endothelial vessels in interfollicular region. Twinning, in which two or more germinal centers are combined and surrounded by lymphocytes in the mantle zone was observed in two cases with lollipop pattern at the same time. All the cases underwent complete surgical resection, the median follow-up time was 48 months(26, 84), both of them had good prognosis. Conclusion:Most cases of unicentric type CD in children are diagnosed late, which is clinical showed by painless lymphadenopathy. The most common pathological type is hyaline vascular. The overall prognosis of surgical treatment was good.


Subject(s)
Castleman Disease , Neck , Humans , Castleman Disease/pathology , Male , Female , Child , Retrospective Studies , Cross-Sectional Studies , Prognosis , Adolescent , Germinal Center/pathology , Child, Preschool
3.
Front Immunol ; 15: 1396808, 2024.
Article in English | MEDLINE | ID: mdl-39136032

ABSTRACT

Introduction: Tertiary lymphoid structures (TLSs) are analogues of secondary lymphoid organs that contain various immune cells. The spatial distribution, maturation and composition of TLSs have differential effects on prognosis, and the roles of TLSs in gastric adenocarcinoma (GA) have not been revealed. Methods: Thus, we evaluated the prognostic value of TLSs in GA through analysis of bulk RNA sequencing(RNA-seq) data from public databases and validated our findings in tumour samples from the Fudan University Shanghai Cancer Center (FUSCC) cohort. The spatial distribution,maturation, and composition of TLSs in GA were analysed by reviewing H&E-stained sections and by multiplex immunofluorescence (mIF) staining. Results: We found that TLSs, especially TLSs with germinal centres (GCs) and TLSs located in the invasive margin (IM), were correlated with prolonged overall survival (OS). Second, analysis of public RNA-seq data showed that high dendritic cell (DC) scores were a favourable prognostic factor in GA patients with high scores for both TLSs and GCs. In the FUSCC cohort, DC-LAMP+ DCs weresignificantly enriched in IM-TLSs with GCs, suggesting a potential correlation between the tumour immune activation milieu and the DC abundance. Third, compared to that in TLSs without GCs, the proportion of FOXP3+CD8+ Treg cells was significantly decreased in IM-TLSs with GCs, and the percentage of PD1+CD20+ B cells was significantly increased in TLSs with GCs. Discussion: Our results demonstrate that the spatial arrangement and maturation of TLSs significantly affect prognosis and indicate that TLSs could be a new additional factor for histopathological evaluation.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Tertiary Lymphoid Structures , Humans , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Stomach Neoplasms/genetics , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/pathology , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Prognosis , Male , Female , Middle Aged , Dendritic Cells/immunology , Aged , Germinal Center/immunology , Germinal Center/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Tumor Microenvironment/immunology
4.
Sci Rep ; 14(1): 17767, 2024 08 01.
Article in English | MEDLINE | ID: mdl-39090233

ABSTRACT

The germinal centers (GCs) are structure found within secondary lymphoid organs and are important for the antibody-producing response against foreign antigens. In GCs, antigen-specific B cells proliferate intensely, inducing immunoglobulin class switching. Recent studies have shown that GCs are also an important site for class switching to IgE, which is implicated in allergy. However, the mechanisms by which IgE production is regulated in GCs remain unclear. Here, we found impairment in IgE-specific production and a reduction of GC B cells after immunization in mice deficient in the Aps/Sh2b2 gene encoding the Lnk/Sh2b family adaptor protein Aps. GC B cells express higher levels of the Aps gene than non-GC B cells, and cell death of Aps-/- GC B cells is enhanced compared to wild-type GC B cells. An in vitro culture system with purified Aps-/- B cells induced the same level of IgE production and frequencies of IgE+ B cells as wild-type B cells. We found that Aps deficiency in B cells resulted in augmented depletion of IgE+ blasts by B cell receptor crosslinking with anti-CD79b antibodies compared to wild-type IgE+ cells. These results suggest that Aps regulates IgE production by controlling the survival of GC B cells and IgE+ plasma cells and may serve as a potential therapeutic target to control IgE production.


Subject(s)
Adaptor Proteins, Signal Transducing , B-Lymphocytes , Cell Survival , Germinal Center , Immunoglobulin E , Animals , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Mice, Inbred C57BL , Mice, Knockout , src Homology Domains
5.
J Med Virol ; 96(8): e29851, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39132689

ABSTRACT

Here, we performed single-cell RNA sequencing of S1 and receptor binding domain protein-specific B cells from convalescent COVID-19 patients with different clinical manifestations. This study aimed to evaluate the role and developmental pathway of atypical memory B cells (MBCs) in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The results revealed a proinflammatory signature across B cell subsets associated with disease severity, as evidenced by the upregulation of genes such as GADD45B, MAP3K8, and NFKBIA in critical and severe individuals. Furthermore, the analysis of atypical MBCs suggested a developmental pathway similar to that of conventional MBCs through germinal centers, as indicated by the expression of several genes involved in germinal center processes, including CXCR4, CXCR5, BCL2, and MYC. Additionally, the upregulation of genes characteristic of the immune response in COVID-19, such as ZFP36 and DUSP1, suggested that the differentiation and activation of atypical MBCs may be influenced by exposure to SARS-CoV-2 and that these genes may contribute to the immune response for COVID-19 recovery. Our study contributes to a better understanding of atypical MBCs in COVID-19 and the role of other B cell subsets across different clinical manifestations.


Subject(s)
COVID-19 , Memory B Cells , SARS-CoV-2 , Single-Cell Analysis , Humans , COVID-19/immunology , COVID-19/virology , COVID-19/genetics , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Memory B Cells/immunology , Male , Adult , Female , Middle Aged , Gene Expression Profiling , Transcriptome , Germinal Center/immunology , B-Lymphocytes/immunology , Aged
6.
Sci Immunol ; 9(98): eadd4874, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39121196

ABSTRACT

Dedicator of cytokinesis 8 (DOCK8) immunodeficiency syndrome is characterized by a failure of the germinal center response, a process involving the proliferation and positive selection of antigen-specific B cells. Here, we describe how DOCK8-deficient B cells are blocked at a light-zone checkpoint in the germinal centers of immunized mice, where they are unable to respond to T cell-dependent survival and selection signals and consequently differentiate into plasma cells or memory B cells. Although DOCK8-deficient B cells can acquire and present antigen to initiate activation of cognate T cells, integrin up-regulation, B cell-T cell conjugate formation, and costimulation are insufficient for sustained B cell and T cell activation when antigen availability is limited. Our findings provide an explanation for the failure of the humoral response in DOCK8 immunodeficiency syndrome and insight into how the level of available antigen modulates B cell-T cell cross-talk to fine-tune humoral immune responses and immunological memory.


Subject(s)
B-Lymphocytes , Guanine Nucleotide Exchange Factors , Mice, Inbred C57BL , T-Lymphocytes , Animals , Guanine Nucleotide Exchange Factors/immunology , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , B-Lymphocytes/immunology , Mice , T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Mice, Knockout , Antigens/immunology , Germinal Center/immunology
7.
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
8.
Sci Rep ; 14(1): 15783, 2024 07 09.
Article in English | MEDLINE | ID: mdl-38982122

ABSTRACT

Peyer's patches (PPs), which contain an abundance of B and T cells, play a key role in inducing pivotal immune responses in the intestinal tract. PPs are defined as aggregated lymph follicles, which consist of multiple lymph follicles (LFs) that may interact with each other in a synergistic manner. LFs are thought to be spherical in shape; however, the characteristics of their structure are not fully understood. To elucidate changes in the structure of PPs as individuals grow, we generated serial 2D sections from entire PPs harvested from mice at 2, 4, and 10 weeks of age and performed a 3D analysis using a software, Amira. Although the number of LFs in PPs was not changed throughout the experiment, the volume and surface area of LFs increased significantly, indicating that LFs in PPs develop continuously by recruiting immune cells, even after weaning. In response to the dramatic changes in the intestinal environment after weaning, the development of germinal centers (GCs) in LFs was observed at 4 and 10 weeks (but not 2 weeks) of age. In addition, GCs gradually began to form away from the center of LFs and close to the muscle layer where export lymphatic vessels develop. Importantly, each LF was joined to the adjacent LF; this feature was observed even in preweaning nonactivated PPs. These results suggest that PPs may have a unique organization and structure that enhance immune functions, allowing cells in LFs to have free access to adjacent LFs and egress smoothly from PPs to the periphery upon stimulation after weaning.


Subject(s)
Peyer's Patches , Weaning , Animals , Peyer's Patches/immunology , Mice , Germinal Center/immunology , B-Lymphocytes/immunology , Intercellular Junctions
9.
Cell Rep ; 43(7): 114454, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990721

ABSTRACT

Memory B cells (MBCs) are essential for humoral immunological memory and can emerge during both the pre-germinal center (GC) and GC phases. However, the transcription regulators governing MBC development remain poorly understood. Here, we report that the transcription regulator Notch2 is highly expressed in MBCs and their precursors at the pre-GC stage and required for MBC development without influencing the fate of GC and plasma cells. Mechanistically, Notch2 signaling promotes the expression of complement receptor CD21 and augments B cell receptor (BCR) signaling. Reciprocally, BCR activation up-regulates Notch2 surface expression in activated B cells via a translation-dependent mechanism. Intriguingly, Notch2 is dispensable for GC-derived MBC formation. In summary, our findings establish Notch2 as a pivotal transcription regulator orchestrating MBC development through the reciprocal enforcement of BCR signaling during the pre-GC phase and suggest that the generation of GC-independent and -dependent MBCs is governed by distinct transcriptional mechanisms.


Subject(s)
Lymphocyte Activation , Memory B Cells , Receptor, Notch2 , Receptors, Antigen, B-Cell , Signal Transduction , Animals , Receptor, Notch2/metabolism , Receptor, Notch2/genetics , Receptors, Antigen, B-Cell/metabolism , Mice , Memory B Cells/metabolism , Memory B Cells/immunology , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Germinal Center/immunology , Germinal Center/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Immunologic Memory , Receptors, Complement 3d/metabolism
10.
Front Immunol ; 15: 1406138, 2024.
Article in English | MEDLINE | ID: mdl-38975334

ABSTRACT

Heterologous prime-boost has broken the protective immune response bottleneck of the COVID-19 vaccines. however, the underlying mechanisms have not been fully elucidated. Here, we investigated antibody responses and explored the response of germinal center (GC) to priming with inactivated vaccines and boosting with heterologous adenoviral-vectored vaccines or homologous inactivated vaccines in mice. Antibody responses were dramatically enhanced by both boosting regimens. Heterologous immunization induced more robust GC activation, characterized by increased Tfh cell populations and enhanced helper function. Additionally, increased B-cell activation and antibody production were observed in a heterologous regimen. Libra-seq was used to compare the differences of S1-, S2- and NTD-specific B cells between homologous and heterologous vaccination, respectively. S2-specific CD19+ B cells presented increased somatic hypermutations (SHMs), which were mainly enriched in plasma cells. Moreover, a heterologous booster dose promoted the clonal expansion of B cells specific to S2 and NTD regions. In conclusion, the functional role of Tfh and B cells following SARS-CoV-2 heterologous vaccination may be important for modulating antibody responses. These findings provide new insights for the development of SARS-CoV-2 vaccines that induce more robust antibody response.


Subject(s)
Antibodies, Viral , Antibody Formation , B-Lymphocytes , COVID-19 Vaccines , COVID-19 , Germinal Center , Immunization, Secondary , SARS-CoV-2 , T Follicular Helper Cells , Animals , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , B-Lymphocytes/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Mice , COVID-19/immunology , COVID-19/prevention & control , T Follicular Helper Cells/immunology , Germinal Center/immunology , Antibody Formation/immunology , Female , Somatic Hypermutation, Immunoglobulin , Vaccination , Mice, Inbred BALB C , Humans , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics
11.
Immunity ; 57(7): 1454-1456, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38986440

ABSTRACT

The association of tertiary lymphoid structures (TLSs) with survival and immunotherapy response brought B cells to center stage. In a pan-cancer B cells atlas in Science, Ma et al. show that germinal center reaction generating anti-tumor antibody-secreting cells (ASCs) from B memory cells in mature TLSs co-exist in tumors with extra-follicular reaction generating auto-reactive ASCs from memory B cells in immature TLSs.


Subject(s)
B-Lymphocytes , Germinal Center , Neoplasms , Humans , B-Lymphocytes/immunology , Germinal Center/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Tertiary Lymphoid Structures/immunology , Memory B Cells/immunology , Immunotherapy/methods
12.
Immunol Cell Biol ; 102(6): 463-466, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38946158

ABSTRACT

In this article for the Highlight of 2023 series, we discuss recent advances in the fundamental biology of the germinal center response. These discoveries provide important insights as to how the germinal center contributes to protection against infection, and also highlights opportunities for future vaccine development.


Subject(s)
Germinal Center , Animals , Humans , B-Lymphocytes/immunology , Germinal Center/immunology , Vaccine Development , Vaccines/immunology
13.
Adv Exp Med Biol ; 1459: 53-77, 2024.
Article in English | MEDLINE | ID: mdl-39017839

ABSTRACT

BOB.1/OBF.1 is a transcriptional coactivator involved in octamer-dependent transcription. Thereby, BOB.1/OBF.1 is involved in the transcriptional regulation of genes important for lymphocyte physiology. BOB.1/OBF.1-deficient mice reveal multiple B- and T-cell developmental defects. The most prominent defect of these mice is the complete absence of germinal centers (GCs) resulting in severely impaired T-cell-dependent immune responses. In humans, BOB.1/OBF.1 is associated with several autoimmune and inflammatory diseases but also linked to liquid and solid tumors. Although its role for B-cell development is relatively well understood, its exact role for the GC reaction and T-cell biology has long been unclear. Here, the contribution of BOB.1/OBF.1 for B-cell maturation is summarized, and recent findings regarding its function in GC B- as well as in various T-cell populations are discussed. Finally, a detailed perspective on how BOB.1/OBF.1 contributes to different pathologies is provided.


Subject(s)
Adaptive Immunity , B-Lymphocytes , T-Lymphocytes , Trans-Activators , Animals , Humans , Adaptive Immunity/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Trans-Activators/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Mice
14.
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
15.
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
16.
Nat Commun ; 15(1): 6421, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080316

ABSTRACT

The rodent-borne Andes virus (ANDV) causes a severe disease in humans. We developed an ANDV mRNA vaccine based on the M segment of the viral genome, either with regular uridine (U-mRNA) or N1-methylpseudouridine (m1Ψ-mRNA). Female mice immunized by m1Ψ-mRNA developed slightly greater germinal center (GC) responses than U-mRNA-immunized mice. Single cell RNA and BCR sequencing of the GC B cells revealed similar levels of activation, except an additional cluster of cells exhibiting interferon response in animals vaccinated with U-mRNA but not m1Ψ-mRNA. Similar immunoglobulin class-switching and somatic hypermutations were observed in response to the vaccines. Female Syrian hamsters were immunized via a prime-boost regimen with two doses of each vaccine. The titers of glycoprotein-binding antibodies were greater for U-mRNA construct than for m1Ψ-mRNA construct; however, the titers of ANDV-neutralizing antibodies were similar. Vaccinated animals were challenged with a lethal dose of ANDV, along with a naïve control group. All control animals and two animals vaccinated with a lower dose of m1Ψ-mRNA succumbed to infection whereas other vaccinated animals survived without evidence of virus replication. The data demonstrate the development of a protective vaccine against ANDV and the lack of a substantial effect of m1Ψ modification on immunogenicity and protection in rodents.


Subject(s)
Mesocricetus , Uridine , Viral Vaccines , Animals , Female , Mice , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/immunology , Antibodies, Viral/immunology , Orthohantavirus/immunology , Orthohantavirus/genetics , Antibodies, Neutralizing/immunology , Germinal Center/immunology , Pseudouridine/immunology , Cricetinae , mRNA Vaccines , Hemorrhagic Fever, American/prevention & control , Hemorrhagic Fever, American/immunology , Hemorrhagic Fever, American/virology , RNA, Viral/genetics , RNA, Viral/immunology , B-Lymphocytes/immunology , Humans , Vaccine Development
17.
J Clin Invest ; 134(12)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38950333

ABSTRACT

Ectopic lymphoid structures (ELSs) in the rheumatoid synovial joints sustain autoreactivity against locally expressed autoantigens. We recently identified recombinant monoclonal antibodies (RA-rmAbs) derived from single, locally differentiated rheumatoid arthritis (RA) synovial B cells, which specifically recognize fibroblast-like synoviocytes (FLSs). Here, we aimed to identify the specificity of FLS-derived autoantigens fueling local autoimmunity and the functional role of anti-FLS antibodies in promoting chronic inflammation. A subset of anti-FLS RA-rmAbs reacting with a 60 kDa band from FLS extracts demonstrated specificity for HSP60 and partial cross-reactivity to other stromal autoantigens (i.e., calreticulin/vimentin) but not to citrullinated fibrinogen. Anti-FLS RA-rmAbs, but not anti-neutrophil extracellular traps rmAbs, exhibited pathogenic properties in a mouse model of collagen-induced arthritis. In patients, anti-HSP60 antibodies were preferentially detected in RA versus osteoarthritis (OA) synovial fluid. Synovial HSPD1 and CALR gene expression analyzed using bulk RNA-Seq and GeoMx-DSP closely correlated with the lympho-myeloid RA pathotype, and HSP60 protein expression was predominantly observed around ELS. Moreover, we observed a significant reduction in synovial HSP60 gene expression followed B cell depletion with rituximab that was strongly associated with the treatment response. Overall, we report that synovial stromal-derived autoantigens are targeted by pathogenic autoantibodies and are associated with specific RA pathotypes, with potential value for patient stratification and as predictors of the response to B cell-depleting therapies.


Subject(s)
Arthritis, Rheumatoid , Autoantigens , Chaperonin 60 , Germinal Center , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Animals , Humans , Mice , Autoantigens/immunology , Autoantigens/genetics , Germinal Center/immunology , Germinal Center/pathology , Chaperonin 60/immunology , Chaperonin 60/genetics , Autoantibodies/immunology , Autoimmunity , Male , Synoviocytes/immunology , Synoviocytes/pathology , Synoviocytes/metabolism , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Female , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/pathology
18.
Nat Immunol ; 25(8): 1432-1444, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969872

ABSTRACT

Memory B cells (MBCs) differentiate into plasma cells (PCs) or germinal centers (GCs) upon antigen recall. How this decision is programmed is not understood. We found that the relative strength between two antagonistic transcription factors, B lymphocyte-induced maturation protein 1 (BLIMP1) and BTB domain and CNC homolog 2 (BACH2), progressively increases in favor of BLIMP1 in antigen-responding B cells through the course of primary responses. MBC subsets that preferentially produce secondary GCs expressed comparatively higher BACH2 but lower BLIMP1 than those predisposed for PC development. Skewing the BLIMP1-BACH2 balance in otherwise fate-predisposed MBC subsets could switch their fate preferences. Underlying the changing BLIMP1-over-BACH2 balance, we observed progressively increased accessibilities at chromatin loci that are specifically opened in PCs, particularly those that contain interferon-sensitive response elements (ISREs) and are controlled by interferon regulatory factor 4 (IRF4). IRF4 is upregulated by B cell receptor, CD40 or innate receptor signaling and it induces graded levels of PC-specifying epigenetic imprints according to the strength of stimulation. By analyzing history-stamped GC B cells, we found progressively increased chromatin accessibilities at PC-specific, IRF4-controlled gene loci over time. Therefore, the cumulative stimulation history of B cells is epigenetically recorded in an IRF4-dependent manner, determines the relative strength between BLIMP1 and BACH2 in individual MBCs and dictates their probabilities to develop into GCs or PCs upon restimulation.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Cell Differentiation , Epigenesis, Genetic , Germinal Center , Immunologic Memory , Interferon Regulatory Factors , Memory B Cells , Plasma Cells , Positive Regulatory Domain I-Binding Factor 1 , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Animals , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Memory B Cells/immunology , Memory B Cells/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Mice, Inbred C57BL , Signal Transduction , Lymphocyte Activation/genetics
19.
Sci Rep ; 14(1): 16970, 2024 07 23.
Article in English | MEDLINE | ID: mdl-39043800

ABSTRACT

B cells and the antibodies they produce are critical in host defense against pathogens and contribute to various immune-mediated diseases. B cells responding to activating signals in vitro release extracellular vesicles (EV) that carry surface antibodies, yet B cell production of EVs that express antibodies and their function in vivo is incompletely understood. Using transgenic mice expressing the Cre recombinase in B cells switching to IgG1 to induce expression of fusion proteins between emerald green fluorescent protein (emGFP) and the EV tetraspanin CD63 as a model, we identify emGFP expression in B cells responding to foreign antigen in vivo and characterize the emGFP+ EVs they release. Our data suggests that emGFP+ germinal center B cells undergoing immunoglobulin class switching to express IgG and their progeny memory B cells and plasma cells, also emGFP+, are sources of circulating antigen-specific IgG+ EVs. Furthermore, using a mouse model of influenza virus infection, we find that IgG+ EVs specific for the influenza hemagglutinin antigen protect against virus infection. In addition, crossing the B cell Cre driver EV reporter mice onto the Nba2 lupus-prone strain revealed increased circulating emGFP+ EVs that expressed surface IgG against nuclear antigens linked to autoimmunity. These data identify EVs loaded with antibodies as a novel route for antibody secretion in B cells that contribute to adaptive immune responses, with important implications for different functions of IgG+ EVs in infection and autoimmunity.


Subject(s)
B-Lymphocytes , Extracellular Vesicles , Immunoglobulin G , Mice, Transgenic , Animals , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Orthomyxoviridae Infections/immunology , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Antigens/immunology , Immunoglobulin Class Switching , Mice, Inbred C57BL , Germinal Center/immunology , Germinal Center/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL