Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.502
Filter
1.
J Exp Med ; 221(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39297882

ABSTRACT

The establishment of long-lasting immunity against pathogens is facilitated by the germinal center (GC) reaction, during which B cells increase their antibody affinity and differentiate into antibody-secreting cells (ASC) and memory cells. These events involve modifications in chromatin packaging that orchestrate the profound restructuring of gene expression networks that determine cell fate. While several chromatin remodelers were implicated in lymphocyte functions, less is known about SMARCA5. Here, using ribosomal pull-down for analyzing translated genes in GC B cells, coupled with functional experiments in mice, we identified SMARCA5 as a key chromatin remodeler in B cells. While the naive B cell compartment remained unaffected following conditional depletion of Smarca5, effective proliferation during B cell activation, immunoglobulin class switching, and as a result GC formation and ASC differentiation were impaired. Single-cell multiomic sequencing analyses revealed that SMARCA5 is crucial for facilitating the transcriptional modifications and genomic accessibility of genes that support B cell activation and differentiation. These findings offer novel insights into the functions of SMARCA5, which can be targeted in various human pathologies.


Subject(s)
B-Lymphocytes , Cell Differentiation , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone , Germinal Center , Animals , Germinal Center/immunology , Germinal Center/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Mice , Mice, Inbred C57BL , Lymphocyte Activation/immunology , Immunoglobulin Class Switching/genetics , Adenosine Triphosphatases
2.
Cell Genom ; 4(9): 100637, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39208795

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) enables comprehensive characterization of the micro-evolutionary processes of B cells during an adaptive immune response, capturing features of somatic hypermutation (SHM) and class switch recombination (CSR). Existing phylogenetic approaches for reconstructing B cell evolution have primarily focused on the SHM process alone. Here, we present tree inference of B cell clonal lineages (TRIBAL), an algorithm designed to optimally reconstruct the evolutionary history of B cell clonal lineages undergoing both SHM and CSR from scRNA-seq data. Through simulations, we demonstrate that TRIBAL produces more comprehensive and accurate B cell lineage trees compared to existing methods. Using real-world datasets, TRIBAL successfully recapitulates expected biological trends in a model affinity maturation system while reconstructing evolutionary histories with more parsimonious class switching than state-of-the-art methods. Thus, TRIBAL significantly improves B cell lineage tracing, useful for modeling vaccine responses, disease progression, and the identification of therapeutic antibodies.


Subject(s)
Algorithms , B-Lymphocytes , Cell Lineage , Single-Cell Analysis , B-Lymphocytes/immunology , Single-Cell Analysis/methods , Cell Lineage/genetics , Humans , Phylogeny , Somatic Hypermutation, Immunoglobulin/genetics , Immunoglobulin Class Switching/genetics , Sequence Analysis, RNA/methods
4.
Expert Rev Mol Diagn ; 24(8): 715-727, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39087770

ABSTRACT

INTRODUCTION: Inborn errors of immunity (IEIs) refer to a heterogeneous category of diseases with defects in the number and/or function of components of the immune system. Immunoglobulin A (IgA) deficiency is the most prevalent IEI characterized by low serum level of IgA and normal serum levels of IgG and/or IgM. Most of the individuals with IgA deficiency are asymptomatic and are only identified through routine laboratory tests. Others may experience a wide range of clinical features including mucosal infections, allergies, and malignancies as the most important features. IgA deficiency is a multi-complex disease, and the exact pathogenesis of it is still unknown. AREAS COVERED: This review compiles recent research on genetic and epigenetic factors that may contribute to the development of IgA deficiency. These factors include defects in B-cell development, IgA class switch recombination, synthesis, secretion, and the long-term survival of IgA switched memory B cells and plasma cells. EXPERT OPINION: A better and more comprehensive understanding of the cellular pathways involved in IgA deficiency could lead to personalized surveillance and potentially curative strategies for affected patients, especially those with severe symptoms.


Subject(s)
IgA Deficiency , Humans , IgA Deficiency/genetics , IgA Deficiency/diagnosis , Immunoglobulin A/blood , Immunoglobulin A/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Genetic Predisposition to Disease , Epigenesis, Genetic , Immunoglobulin Class Switching/genetics
5.
Nat Commun ; 15(1): 7541, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39215025

ABSTRACT

A diverse antibody repertoire is essential for humoral immunity. Antibody diversification requires the introduction of deoxyuridine (dU) mutations within immunoglobulin genes to initiate somatic hypermutation (SHM) and class switch recombination (CSR). dUs are normally recognized and excised by the base excision repair (BER) protein uracil-DNA glycosylase 2 (UNG2). However, FAM72A downregulates UNG2 permitting dUs to persist and trigger SHM and CSR. How FAM72A promotes UNG2 degradation is unknown. Here, we show that FAM72A recruits a C-terminal to LisH (CTLH) E3 ligase complex to target UNG2 for proteasomal degradation. Deficiency in CTLH complex components result in elevated UNG2 and reduced SHM and CSR. Cryo-EM structural analysis reveals FAM72A directly binds to MKLN1 within the CTLH complex to recruit and ubiquitinate UNG2. Our study further suggests that FAM72A hijacks the CTLH complex to promote mutagenesis in cancer. These findings show that FAM72A is an E3 ligase substrate adaptor critical for humoral immunity and cancer development.


Subject(s)
Immunoglobulin Class Switching , Ubiquitin-Protein Ligases , Humans , Animals , Immunoglobulin Class Switching/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Mice , DNA Glycosylases/metabolism , DNA Glycosylases/genetics , HEK293 Cells , Ubiquitination , Somatic Hypermutation, Immunoglobulin/genetics , Mutagenesis , DNA Repair , Proteolysis , Immunity, Humoral , Mice, Inbred C57BL
6.
Immunol Lett ; 269: 106909, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128629

ABSTRACT

Activation-induced cytidine deaminase (AID) is responsible for the initiation of somatic hypermutation (SHM) and class-switch recombination (CSR), which result in antibody affinity maturation and isotype switching, thus producing pathogen-specific antibodies. Chromatin dynamics and accessibility play a significant role in determining AID expression and its targeting. Chromatin remodelers contribute to the accessibility of the chromatin structure, thereby influencing the targeting of AID to Ig genes. Epigenetic modifications, including DNA methylation, histone modifications, and miRNA expression, profoundly impact the regulation of AID and chromatin remodelers targeting Ig genes. Additionally, epigenetic modifications lead to chromatin rearrangement and thereby can change AID expression levels and its preferential targeting to Ig genes. This interplay is symbolized as the ACE phenomenon encapsulates three interconnected aspects: AID, Chromatin remodelers, and Epigenetic modifications. This review emphasizes the importance of understanding the intricate relationship between these aspects to unlock the therapeutic potential of these molecular processes and molecules.


Subject(s)
Antibody Diversity , Chromatin Assembly and Disassembly , Cytidine Deaminase , Epigenesis, Genetic , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Humans , Animals , Antibody Diversity/genetics , Immunoglobulin Class Switching/genetics , DNA Methylation , Somatic Hypermutation, Immunoglobulin , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation
7.
Adv Exp Med Biol ; 1445: 37-46, 2024.
Article in English | MEDLINE | ID: mdl-38967748

ABSTRACT

It is widely acknowledged that immunoglobulins (Igs) are produced solely by B-lineage cells. The Ig gene is created by the rearrangement of a group of gene segments [variable (V), diversity (D), and joining (J) segments rearrangement, or V(D)J recombination], which results in the vast diversity of B cell-derived Ig responsible for recognising various antigens. Ig subsequently undergoes somatic hypermutation (SHM) and class switch recombination (CSR) after exposure to antigens, thus converting the low-affinity IgM to IgG, IgA, or IgE antibodies. IgM and IgD are primarily expressed in naïve B cells that have not been exposed to antigens, they do not undergo somatic hypermutation; hence, their variable region sequences remain the same as those in the germline. In contrast, IgG, IgA, and IgE are expressed in antigen-stimulated memory B cells or plasma cells, and thus, they often possess high-frequency mutations in their variable region sequences. Since the discovery that Ig can be produced by non-B cells, Qiu's group has investigated and compared the genetic characteristics of B cell-derived Ig and non-B cell-derived Ig. These findings demonstrated that non-B cell-derived Ig shares certain similarities with B cell-derived Ig in that the sequence of its constant region is identical to that of B cell-derived Ig, and its variable region is also strictly dependent on the rearrangement of V, D, and J gene segments. Moreover, akin to B cell-derived Ig, the V regions of IgM and IgD are rarely mutated, while IgG, IgA, and IgE produced by cancer cells are frequently mutated. However, the non-B cell-derived Ig V region sequence displays unique characteristics. (1) Unlike the vast diversity of B cell-derived Igs, non-B cell-derived Igs exhibit restricted diversity; cells from the same lineage always select the same V(D)J recombination patterns; (2) Both mRNA and proteins of RAG1/RAG2 recombinase have been detected in Ig positive cancer cell lines and normal tissues. But Ig recombination could also be found in RAG1-/- and RAG2-/- mice, suggesting that they are not necessary for the rearrangement of non-B cell-derived Igs. These features of non-B cell-derived Igs suggest a potentially undiscovered mechanism of V(D)J recombination, ligation, and SHM in non-B cells, which necessitates further investigation with advanced technology in molecular biology.


Subject(s)
B-Lymphocytes , Genes, Immunoglobulin , Animals , Humans , Mice , B-Lymphocytes/immunology , Genes, Immunoglobulin/genetics , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunoglobulins/genetics , Immunoglobulins/immunology , Somatic Hypermutation, Immunoglobulin/genetics
8.
Methods Mol Biol ; 2826: 167-187, 2024.
Article in English | MEDLINE | ID: mdl-39017893

ABSTRACT

Division tracking dyes like Cell Trace Violet (CTV) enable the quantification of cell proliferation, division, and survival kinetics of human naïve B cell responses in vitro. Human naïve B cells exhibit distinct responses to different stimuli, with CpG and anti-Ig inducing a T cell-independent (TI) response, while CD40L and IL-21 promote a T cell-dependent (TD) response that induces isotype switching and differentiation into antibody-secreting cells (ASCs). Both stimulation methods yield valuable insights into the intrinsic programming of B cell health within individuals, making them useful for clinical investigations. For instance, quantitative analysis from these B cell populations could reveal biologically meaningful measurements such as the average number of division rounds and the time to cells' fate. Here, we describe a novel in vitro culture setup for CTV-labelled human naïve B cells and a method for obtaining precise time-based data on proliferation, division-linked isotype switching, and differentiation.


Subject(s)
B-Lymphocytes , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Humans , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Culture Techniques/methods , Kinetics , Lymphocyte Activation , Cells, Cultured , Immunoglobulin Class Switching
9.
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
10.
BMC Biol ; 22(1): 151, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977974

ABSTRACT

BACKGROUND: RNA-DNA hybrids or R-loops are associated with deleterious genomic instability and protective immunoglobulin class switch recombination (CSR). However, the underlying phenomenon regulating the two contrasting functions of R-loops is unknown. Notably, the underlying mechanism that protects R-loops from classic RNase H-mediated digestion thereby promoting persistence of CSR-associated R-loops during CSR remains elusive. RESULTS: Here, we report that during CSR, R-loops formed at the immunoglobulin heavy (IgH) chain are modified by ribose 2'-O-methylation (2'-OMe). Moreover, we find that 2'-O-methyltransferase fibrillarin (FBL) interacts with activation-induced cytidine deaminase (AID) associated snoRNA aSNORD1C to facilitate the 2'-OMe. Moreover, deleting AID C-terminal tail impairs its association with aSNORD1C and FBL. Disrupting FBL, AID or aSNORD1C expression severely impairs 2'-OMe, R-loop stability and CSR. Surprisingly, FBL, AID's interaction partner and aSNORD1C promoted AID targeting to the IgH locus. CONCLUSION: Taken together, our results suggest that 2'-OMe stabilizes IgH-associated R-loops to enable productive CSR. These results would shed light on AID-mediated CSR and explain the mechanism of R-loop-associated genomic instability.


Subject(s)
Cytidine Deaminase , Immunoglobulin Class Switching , R-Loop Structures , Immunoglobulin Class Switching/genetics , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/chemistry , Animals , Mice , Methylation , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Recombination, Genetic , RNA/metabolism , RNA/genetics
11.
Front Immunol ; 15: 1429909, 2024.
Article in English | MEDLINE | ID: mdl-39081315

ABSTRACT

Previous studies have demonstrated the efficacy and feasibility of an anti-viral vaccine strategy that takes advantage of pre-existing CD4+ helper T (Th) cells induced by Mycobacterium bovis bacille Calmette-Guérin (BCG) vaccination. This strategy uses immunization with recombinant fusion proteins comprised of a cell surface expressed viral antigen, such as a viral envelope glycoprotein, engineered to contain well-defined BCG Th cell epitopes, thus rapidly recruiting Th cells induced by prior BCG vaccination to provide intrastructural help to virus-specific B cells. In the current study, we show that Th cells induced by BCG were localized predominantly outside of germinal centers and promoted antibody class switching to isotypes characterized by strong Fc receptor interactions and effector functions. Furthermore, BCG vaccination also upregulated FcγR expression to potentially maximize antibody-dependent effector activities. Using a mouse model of Ebola virus (EBOV) infection, this vaccine strategy provided sustained antibody levels with strong IgG2c bias and protection against lethal challenge. This general approach can be easily adapted to other viruses, and may be a rapid and effective method of immunization against emerging pandemics in populations that routinely receive BCG vaccination.


Subject(s)
Antibodies, Viral , BCG Vaccine , Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Mice , BCG Vaccine/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/immunology , Ebola Vaccines/immunology , Ebola Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , T-Lymphocytes, Helper-Inducer/immunology , Vaccination/methods , Mice, Inbred C57BL , Female , Humans , Disease Models, Animal , Receptors, IgG/immunology , Vaccine Development , Immunoglobulin Class Switching , Immunization
12.
Nat Immunol ; 25(8): 1489-1506, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38918608

ABSTRACT

Humanized mice are limited in terms of modeling human immunity, particularly with regards to antibody responses. Here we constructed a humanized (THX) mouse by grafting non-γ-irradiated, genetically myeloablated KitW-41J mutant immunodeficient pups with human cord blood CD34+ cells, followed by 17ß-estradiol conditioning to promote immune cell differentiation. THX mice reconstitute a human lymphoid and myeloid immune system, including marginal zone B cells, germinal center B cells, follicular helper T cells and neutrophils, and develop well-formed lymph nodes and intestinal lymphoid tissue, including Peyer's patches, and human thymic epithelial cells. These mice have diverse human B cell and T cell antigen receptor repertoires and can mount mature T cell-dependent and T cell-independent antibody responses, entailing somatic hypermutation, class-switch recombination, and plasma cell and memory B cell differentiation. Upon flagellin or a Pfizer-BioNTech coronavirus disease 2019 (COVID-19) mRNA vaccination, THX mice mount neutralizing antibody responses to Salmonella or severe acute respiratory syndrome coronavirus 2 Spike S1 receptor-binding domain, with blood incretion of human cytokines, including APRIL, BAFF, TGF-ß, IL-4 and IFN-γ, all at physiological levels. These mice can also develop lupus autoimmunity after pristane injection. By leveraging estrogen activity to support human immune cell differentiation and maturation of antibody responses, THX mice provide a platform to study the human immune system and to develop human vaccines and therapeutics.


Subject(s)
Antibodies, Neutralizing , Immunoglobulin Class Switching , Animals , Humans , Mice , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , SARS-CoV-2/immunology , COVID-19/immunology , Antibodies, Viral/immunology , Somatic Hypermutation, Immunoglobulin , Cell Differentiation/immunology
13.
Biom J ; 66(4): e2300171, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38785212

ABSTRACT

Statistical and machine learning methods have proved useful in many areas of immunology. In this paper, we address for the first time the problem of predicting the occurrence of class switch recombination (CSR) in B-cells, a problem of interest in understanding antibody response under immunological challenges. We propose a framework to analyze antibody repertoire data, based on clonal (CG) group representation in a way that allows us to predict CSR events using CG level features as input. We assess and compare the performance of several predicting models (logistic regression, LASSO logistic regression, random forest, and support vector machine) in carrying out this task. The proposed approach can obtain an unweighted average recall of 71 % $71\%$ with models based on variable region descriptors and measures of CG diversity during an immune challenge and, most notably, before an immune challenge.


Subject(s)
B-Lymphocytes , Immunoglobulin Class Switching , B-Lymphocytes/immunology , Animals , Biometry/methods , Recombination, Genetic , Antibodies/immunology , Mice , Humans
14.
Adv Immunol ; 161: 127-164, 2024.
Article in English | MEDLINE | ID: mdl-38763700

ABSTRACT

Activation induced cytidine deaminase (AID) is a key element of the adaptive immune system, required for immunoglobulin isotype switching and affinity maturation of B-cells as they undergo the germinal center (GC) reaction in peripheral lymphoid tissue. The inherent DNA damaging activity of this enzyme can also have off-target effects in B-cells, producing lymphomagenic chromosomal translocations that are characteristic features of various classes of non-Hodgkin B-cell lymphoma (B-NHL), and generating oncogenic mutations, so-called aberrant somatic hypermutation (aSHM). Additionally, AID has been found to affect gene expression through demethylation as well as altered interactions between gene regulatory elements. These changes have been most thoroughly studied in B-NHL arising from GC B-cells. Here, we describe the most common classes of GC-derived B-NHL and explore the consequences of on- and off-target AID activity in B and plasma cell neoplasms. The relationships between AID expression, including effects of infection and other exposures/agents, mutagenic activity and lymphoma biology are also discussed.


Subject(s)
B-Lymphocytes , Cytidine Deaminase , Germinal Center , Lymphoma, B-Cell , Humans , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Germinal Center/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Animals , B-Lymphocytes/immunology , Somatic Hypermutation, Immunoglobulin , Gene Expression Regulation, Neoplastic , Immunoglobulin Class Switching
15.
Proc Natl Acad Sci U S A ; 121(22): e2314619121, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38776375

ABSTRACT

Humoral immunity depends on the germinal center (GC) reaction where B cells are tightly controlled for class-switch recombination and somatic hypermutation and finally generated into plasma and memory B cells. However, how protein SUMOylation regulates the process of the GC reaction remains largely unknown. Here, we show that the expression of SUMO-specific protease 1 (SENP1) is up-regulated in GC B cells. Selective ablation of SENP1 in GC B cells results in impaired GC dark and light zone organization and reduced IgG1-switched GC B cells, leading to diminished production of class-switched antibodies with high-affinity in response to a TD antigen challenge. Mechanistically, SENP1 directly binds to Paired box protein 5 (PAX5) to mediate PAX5 deSUMOylation, sustaining PAX5 protein stability to promote the transcription of activation-induced cytidine deaminase. In summary, our study uncovers SUMOylation as an important posttranslational mechanism regulating GC B cell response.


Subject(s)
B-Lymphocytes , Cysteine Endopeptidases , Germinal Center , PAX5 Transcription Factor , Sumoylation , Germinal Center/immunology , Germinal Center/metabolism , PAX5 Transcription Factor/metabolism , PAX5 Transcription Factor/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Mice , Immunoglobulin Class Switching , Humans , Cytidine Deaminase/metabolism , Cytidine Deaminase/genetics , Immunity, Humoral , Mice, Inbred C57BL
16.
Front Immunol ; 15: 1405022, 2024.
Article in English | MEDLINE | ID: mdl-38799442

ABSTRACT

Immunodeficiency, centromeric instability, and facial anomalies (ICF) syndrome, a rare autosomal recessive disorder, manifests with hypoglobulinemia and chromosomal instability accompanied by DNA hypomethylation. Pathological variants in the DNMT3B, ZBTB24, CDCA7, or HELLS genes underlie its etiology. Activated lymphocytes from patients often display distinctive multiradial chromosomes fused via pericentromeric regions. Recent studies have provided deeper insights into how pathological variants in ICF-related proteins cause DNA hypomethylation and chromosome instability. However, the understanding of the molecular pathogenesis underlying immunodeficiency is still in its nascent stages. In the past half-decade, the roles of CDCA7, HELLS, and ZBTB24 in classical non-homologous end joining during double-strand DNA break repair and immunoglobulin class-switch recombination (CSR) have been unveiled. Nevertheless, given the decreased all classes of immunoglobulins in most patients, CSR deficiency alone cannot fully account for the immunodeficiency. The latest finding showing dysregulation of immunoglobulin signaling may provide a clue to understanding the immunodeficiency mechanism. While less common, a subgroup of patients exhibits T-cell abnormalities alongside B-cell anomalies, including reduced regulatory T-cells and increased effector memory T- and follicular helper T-cells. The dysregulation of immunoglobulin signaling in B-cells, the imbalance in T-cell subsets, and/or satellite RNA-mediated activation of innate immune response potentially explain autoimmune manifestations in a subset of patients. These findings emphasize the pivotal roles of ICF-related proteins in both B- and T-cell functions. ICF syndrome studies have illuminated many fundamental mechanisms. Further investigations will certainly continue to unveil additional mechanisms and their interplay.


Subject(s)
DNA Repair , Epigenesis, Genetic , Immunologic Deficiency Syndromes , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , DNA Methylation , Animals , Immunoglobulin Class Switching/genetics , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/diagnosis
17.
Cell Rep ; 43(5): 114194, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38735043

ABSTRACT

Class switch recombination (CSR) diversifies the effector functions of antibodies and involves complex regulation of transcription and DNA damage repair. Here, we show that the deubiquitinase USP7 promotes CSR to immunoglobulin A (IgA) and suppresses unscheduled IgG switching in mature B cells independent of its role in DNA damage repair, but through modulating switch region germline transcription. USP7 depletion impairs Sα transcription, leading to abnormal activation of Sγ germline transcription and increased interaction with the CSR center via loop extrusion for unscheduled IgG switching. Rescue of Sα transcription by transforming growth factor ß (TGF-ß) in USP7-deleted cells suppresses Sγ germline transcription and prevents loop extrusion toward IgG CSR. Mechanistically, USP7 protects transcription factor RUNX3 from ubiquitination-mediated degradation to promote Sα germline transcription. Our study provides evidence for active transcription serving as an anchor to impede loop extrusion and reveals a functional interplay between USP7 and TGF-ß signaling in promoting RUNX3 expression for efficient IgA CSR.


Subject(s)
Core Binding Factor Alpha 3 Subunit , Immunoglobulin A , Immunoglobulin Class Switching , Transcriptional Activation , Ubiquitin-Specific Peptidase 7 , Animals , Humans , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Immunoglobulin A/metabolism , Immunoglobulin G/metabolism , Immunoglobulin G/immunology , Mice, Inbred C57BL , Protein Stability , Transforming Growth Factor beta/metabolism , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Ubiquitination
19.
Nucleic Acids Res ; 52(8): 4422-4439, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38567724

ABSTRACT

Efficient repair of DNA double-strand breaks in the Ig heavy chain gene locus is crucial for B-cell antibody class switch recombination (CSR). The regulatory dynamics of the repair pathway direct CSR preferentially through nonhomologous end joining (NHEJ) over alternative end joining (AEJ). Here, we demonstrate that the histone acetyl reader BRD2 suppresses AEJ and aberrant recombination as well as random genomic sequence capture at the CSR junctions. BRD2 deficiency impairs switch (S) region synapse, optimal DNA damage response (DDR), and increases DNA break end resection. Unlike BRD4, a similar bromodomain protein involved in NHEJ and CSR, BRD2 loss does not elevate RPA phosphorylation and R-loop formation in the S region. As BRD2 stabilizes the cohesion loader protein NIPBL in the S regions, the loss of BRD2 or NIPBL shows comparable deregulation of S-S synapsis, DDR, and DNA repair pathway choice during CSR. This finding extends beyond CSR, as NIPBL and BRD4 have been linked to Cornelia de Lange syndrome, a developmental disorder exhibiting defective NHEJ and Ig isotype switching. The interplay between these proteins sheds light on the intricate mechanisms governing DNA repair and immune system functionality.


Subject(s)
Bromodomain Containing Proteins , DNA End-Joining Repair , Immunoglobulin Class Switching , Transcription Factors , Animals , Humans , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bromodomain Containing Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA Repair , Immunoglobulin Class Switching/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Recombination, Genetic , Transcription Factors/metabolism
20.
EMBO J ; 43(10): 1947-1964, 2024 May.
Article in English | MEDLINE | ID: mdl-38605225

ABSTRACT

Transcription factors BACH2 and IRF4 are both essential for antibody class-switch recombination (CSR) in activated B lymphocytes, while they oppositely regulate the differentiation of plasma cells (PCs). Here, we investigated how BACH2 and IRF4 interact during CSR and plasma-cell differentiation. We found that BACH2 organizes heterochromatin formation of target gene loci in mouse splenic B cells, including targets of IRF4 activation such as Aicda, an inducer of CSR, and Prdm1, a master plasma-cell regulator. Release of these gene loci from heterochromatin in response to B-cell receptor stimulation was coupled to AKT-mTOR pathway activation. In Bach2-deficient B cells, PC genes' activation depended on IRF4 protein accumulation, without an increase in Irf4 mRNA. Mechanistically, a PU.1-IRF4 heterodimer in activated B cells promoted BACH2 function by inducing gene expression of Bach2 and Pten, a negative regulator of AKT signaling. Elevated AKT activity in Bach2-deficient B cells resulted in IRF4 protein accumulation. Thus, BACH2 and IRF4 mutually modulate the activity of each other, and BACH2 inhibits PC differentiation by both the repression of PC genes and the restriction of IRF4 protein accumulation.


Subject(s)
Basic-Leucine Zipper Transcription Factors , Cell Differentiation , Interferon Regulatory Factors , Plasma Cells , Animals , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/cytology , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Heterochromatin/metabolism , Heterochromatin/genetics , Immunoglobulin Class Switching/genetics , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Plasma Cells/metabolism , Plasma Cells/immunology , Plasma Cells/cytology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Trans-Activators/metabolism , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL