Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Sci Adv ; 9(17): eade8928, 2023 04 28.
Article in English | MEDLINE | ID: mdl-37115935

ABSTRACT

Maturation of antibody responses entails somatic hypermutation (SHM), class-switch DNA recombination (CSR), plasma cell differentiation, and generation of memory B cells, and it is thought to require T cell help. We showed that B cell Toll-like receptor 4 (TLR4)-B cell receptor (BCR) (receptor for antigen) coengagement by 4-hydroxy-3-nitrophenyl acetyl (NP)-lipopolysaccharide (LPS) (Escherichia coli lipid A polysaccharide O-antigen) or TLR5-BCR coengagement by Salmonella flagellin induces mature antibody responses to NP and flagellin in Tcrß-/-Tcrδ-/- and NSG/B mice. TLR-BCR coengagement required linkage of TLR and BCR ligands, "linked coengagement." This induced B cell CSR/SHM, germinal center-like differentiation, clonal expansion, intraconal diversification, plasma cell differentiation, and an anamnestic antibody response. In Tcrß-/-Tcrδ-/- mice, linked coengagement of TLR4-BCR by LPS or TLR5-BCR by flagellin induced protective antibodies against E. coli or Salmonella Typhimurium. Our findings unveiled a critical role of B cell TLRs in inducing neutralizing antibody responses, including those to microbial pathogens, without T cell help.


Subject(s)
Antibody Formation , Toll-Like Receptor 4 , Mice , Animals , Toll-Like Receptor 4/genetics , Antibodies, Neutralizing , Lipopolysaccharides , Escherichia coli , Flagellin , Toll-Like Receptor 5/genetics , T-Lymphocytes , Receptors, Antigen, B-Cell
2.
Front Immunol ; 13: 882706, 2022.
Article in English | MEDLINE | ID: mdl-35911693

ABSTRACT

Hemichannels (HCs)/gap junctions (GJs) and immunoglobulin (Ig)-like domain-containing proteins (IGLDCPs) are involved in the innate-adaptive immune response independently. Despite of available evidence demonstrating the importance of HCs/GJs and IGLDCPs in initiating, implementing, and terminating the entire immune response, our understanding of their mutual interactions in immunological function remains rudimentary. IGLDCPs include immune checkpoint molecules of the immunoglobulin family expressed in T and B lymphocytes, most of which are cluster of differentiation (CD) antigens. They also constitute the principal components of the immunological synapse (IS), which is formed on the cell surface, including the phagocytic synapse, T cell synapse, B cell synapse, and astrocytes-neuronal synapse. During the three stages of the immune response, namely innate immunity, innate-adaptive immunity, and adaptive immunity, HCs/GJs and IGLDCPs are cross-activated during the entire process. The present review summarizes the current understanding of HC-released immune signaling factors that influence IGLDCPs in regulating innate-adaptive immunity. ATP-induced "eat me" signals released by HCs, as well as CD31, CD47, and CD46 "don't eat me" signaling molecules, trigger initiation of innate immunity, which serves to regulate phagocytosis. Additionally, HC-mediated trogocytosis promotes antigen presentation and amplification. Importantly, HC-mediated CD4+ T lymphocyte activation is critical in the transition of the innate immune response to adaptive immunity. HCs also mediate non-specific transcytosis of antibodies produced by mature B lymphocytes, for instance, IgA transcytosis in ovarian cancer cells, which triggers innate immunity. Further understanding of the interplay between HCs/GJs and IGLDCPs would aid in identifying therapeutic targets that regulate the HC-Ig-like domain immune response, thereby providing a viable treatment strategy for immunological diseases. The present review delineates the clinical immunology-related applications of HC-Ig-like domain cross-activation, which would greatly benefit medical professionals and immunological researchers alike. HCs/GJs and IGLDCPs mediate phagocytosis via ATP; "eat me and don't eat me" signals trigger innate immunity; HC-mediated trogocytosis promotes antigen presentation and amplification in innate-adaptive immunity; HCs also mediate non-specific transcytosis of antibodies produced by mature B lymphocytes in adaptive immunity.


Subject(s)
Adaptive Immunity , Immunity, Innate , Adenosine Triphosphate , Antigens, CD , Gap Junctions , Immunoglobulin Domains
3.
Nat Commun ; 13(1): 980, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35190531

ABSTRACT

In B cells, IgD is expressed together with IgM through alternative splicing of primary VHDJH-Cµ-s-m-Cδ-s-m RNAs, and also through IgD class switch DNA recombination (CSR) via double-strand DNA breaks (DSB) and synapse of Sµ with σδ. How such DSBs are resolved is still unknown, despite our previous report showing that Rad52 effects the 'short-range' microhomology-mediated synapsis of intra-Sµ region DSBs. Here we find that induction of IgD CSR downregulates Zfp318, and promotes Rad52 phosphorylation and recruitment to Sµ and σδ, thereby leading to alternative end-joining (A-EJ)-mediated Sµ-σδ recombination with extensive microhomologies, VHDJH-Cδs transcription and sustained IgD secretion. Rad52 ablation in mouse Rad52-/- B cells aborts IgD CSR in vitro and in vivo and dampens the specific IgD antibody response to OVA. Rad52 knockdown in human B cells also abrogates IgD CSR. Finally, Rad52 phosphorylation is associated with high levels of IgD CSR and anti-nuclear IgD autoantibodies in patients with systemic lupus erythematosus and in lupus-prone mice. Our findings thus show that Rad52 mediates IgD CSR through microhomology-mediated A-EJ in concert with Zfp318 downregulation.


Subject(s)
Immunoglobulin Class Switching/genetics , Immunoglobulin D/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Animals , B-Lymphocytes , DNA-Binding Proteins/metabolism , Female , Gene Knockdown Techniques , Healthy Volunteers , Humans , Male , Mice , Mice, Knockout , Phosphorylation/genetics , Primary Cell Culture , Rad52 DNA Repair and Recombination Protein/genetics , Recombination, Genetic
4.
Front Immunol ; 12: 761450, 2021.
Article in English | MEDLINE | ID: mdl-34868004

ABSTRACT

IgA is the predominant antibody isotype at intestinal mucosae, where it plays a critical role in homeostasis and provides a first line of immune protection. Dysregulation of IgA production, however, can contribute to immunopathology, particularly in kidneys in which IgA deposition can cause nephropathy. Class-switch DNA recombination (CSR) to IgA is directed by TGF-ß signaling, which activates Smad2 and Smad3. Activated Smad2/Smad3 dimers are recruited together with Smad4 to the IgH α locus Iα promoter to activate germline Iα-Cα transcription, the first step in the unfolding of CSR to IgA. Epigenetic factors, such as non-coding RNAs, particularly microRNAs, have been shown to regulate T cells, dendritic cells and other immune elements, as well as modulate the antibody response, including CSR, in a B cell-intrinsic fashion. Here we showed that the most abundant miRNA in resting B cells, miR-146a targets Smad2, Smad3 and Smad4 mRNA 3'UTRs and keeps CSR to IgA in check in resting B cells. Indeed, enforced miR-146a expression in B cells aborted induction of IgA CSR by decreasing Smad levels. By contrast, upon induction of CSR to IgA, as directed by TGF-ß, B cells downregulated miR-146a, thereby reversing the silencing of Smad2, Smad3 and Smad4, which, once expressed, led to recruitment of Smad2, Smad3 and Smad4 to the Iα promoter for activation of germline Iα-Cα transcription. Deletion of miR-146a in miR-146a-/- mice significantly increased circulating levels of steady state total IgA, but not IgM, IgG or IgE, and heightened the specific IgA antibody response to OVA. In miR-146a-/- mice, the elevated systemic IgA levels were associated with increased IgA+ B cells in intestinal mucosae, increased amounts of fecal free and bacteria-bound IgA as well as kidney IgA deposition, a hallmark of IgA nephropathy. Increased germline Iα-Cα transcription and CSR to IgA in miR-146a-/- B cells in vitro proved that miR-146a-induced Smad2, Smad3 and Smad4 repression is B cell intrinsic. The B cell-intrinsic role of miR-146a in the modulation of CSR to IgA was formally confirmed in vivo by construction and OVA immunization of mixed bone marrow µMT/miR-146a-/- chimeric mice. Thus, by inhibiting Smad2, Smad3 and Smad4 expression, miR-146a plays an important and B cell intrinsic role in modulation of CSR to IgA and the IgA antibody response.


Subject(s)
Epigenesis, Genetic , Immunoglobulin A/genetics , Immunoglobulin Class Switching/genetics , MicroRNAs/physiology , Recombination, Genetic , Smad Proteins/physiology , Animals , Down-Regulation , Gastrointestinal Microbiome , Immunoglobulin A/blood , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Smad2 Protein/physiology , Smad3 Protein/physiology , Smad4 Protein/physiology
5.
Front Immunol ; 12: 658048, 2021.
Article in English | MEDLINE | ID: mdl-33953720

ABSTRACT

B cell activation by Tfh cells, i.e., through CD154 engagement of CD40 and IL-21, and survival within GCs are crucial for the T-dependent Ab response. LUBAC, composed of HOIP, SHARPIN, and HOIL-1, catalyzes linear ubiquitination (Linear M1-Ub) to mediate NF-κB activation and cell survival induced by TNF receptor superfamily members, which include CD40. As shown in this study, B cells expressing the Sharpin null mutation cpdm (Sharpincpdm ) could undergo proliferation, CSR, and SHM in response to immunization by a T-dependent Ag, but were defective in survival within GCs, enrichment of a mutation enhancing the BCR affinity, and production of specific Abs. Sharpincpdm B cells stimulated in vitro with CD154 displayed normal proliferation and differentiation, marginally impaired NF-κB activation and survival, but markedly exacerbated death triggered by IL-21. While activating the mitochondria-dependent apoptosis pathway in both Sharpin+/+ and Sharpincpdm B cells, IL-21 induced Sharpincpdm B cells to undergo sustained activation of caspase 9 and caspase 8 of the mitochondria-dependent and independent pathway, respectively, and ultimately caspase 3 in effecting apoptosis. These were associated with loss of the caspase 8 inhibitor cFLIP and reduction in cFLIP Linear M1-Ub, which interferes with cFLIP poly-ubiquitination at Lys48 and degradation. Finally, the viability of Sharpincpdm B cells was rescued by caspase inhibitors but virtually abrogated - together with Linear M1-Ub and cFLIP levels - by a small molecule HOIP inhibitor. Thus, LUBAC controls the cFLIP expression and inhibits the effects of caspase 8 and IL-21-activated caspase 9, thereby suppressing apoptosis of CD40 and IL-21-activated B cells and promoting GC B cell survival.


Subject(s)
Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Antigens/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Interleukins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Antibody Formation/immunology , Apoptosis/drug effects , Biomarkers , Caspase 8/metabolism , Caspase 9 , Cell Communication , Cell Survival , Mice , Mice, Transgenic , Mutation , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Ubiquitins/metabolism
6.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33400692

ABSTRACT

2'3'-cGAMP is known as a nonclassical second messenger and small immune modulator that possesses potent antitumor and antiviral activities via inducing the stimulator of IFN genes-mediated (STING-mediated) signaling pathway. However, its function in regulating type 2 immune responses remains unknown. Therefore, we sought to determine a role of STING activation by 2'3'-cGAMP in type 2 inflammatory reactions in multiple mouse models of eosinophilic asthma. We discovered that 2'3'-cGAMP administration strongly attenuated type 2 lung immunopathology and airway hyperreactivity induced by IL-33 and a fungal allergen, Aspergillus flavus. Mechanistically, upon the respiratory delivery, 2'3'-cGAMP was mainly internalized by alveolar macrophages, in which it activated the STING/IFN regulatory factor 3/type I IFN signaling axis to induce the production of inhibitory factors containing IFN-α, which blocked the IL-33-mediated activation of group 2 innate lymphoid (ILC2) cells in vivo. We further demonstrated that 2'3'-cGAMP directly suppressed the proliferation and function of both human and mouse ILC2 cells in vitro. Taken together, our findings suggest that STING activation by 2'3'-cGAMP in alveolar macrophages and ILC2 cells can negatively regulate type 2 immune responses, implying that the respiratory delivery of 2'3'-cGAMP might be further developed as an alternative strategy for treating type 2 immunopathologic diseases such as eosinophilic asthma.


Subject(s)
Asthma/immunology , Interleukin-33/metabolism , Lymphocytes/immunology , Macrophages, Alveolar/immunology , Membrane Proteins/metabolism , Allergens/administration & dosage , Animals , Aspergillus flavus/immunology , Asthma/metabolism , Asthma/pathology , Disease Models, Animal , Eosinophilia/immunology , Eosinophilia/metabolism , Eosinophilia/pathology , Female , Guanine Nucleotides/administration & dosage , Guanine Nucleotides/immunology , Guanine Nucleotides/metabolism , Humans , Immunity, Innate , In Vitro Techniques , Interleukin-33/administration & dosage , Interleukin-33/genetics , Lymphocytes/pathology , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction
7.
Curr Opin Immunol ; 67: 75-86, 2020 12.
Article in English | MEDLINE | ID: mdl-33176228

ABSTRACT

B cell differentiation driven by microbial antigens leads to production of anti-microbial antibodies, such as those neutralizing viruses, bacteria or bacterial toxin, that are class-switched (IgG and IgA) and somatically hypermutated (maturation of the antibody response) as well as secreted in large volume by plasma cells. Similar features characterize pathogenic antibodies to self-antigens in autoimmunity, reflecting the critical role of class switch DNA recombination (CSR), somatic hypermutation (SHM) and plasma cell differentiation in the generation of antibodies to not only foreign antigens but also self-antigens (autoantibodies). Central to CSR/SHM and plasma cell differentiation are AID, a potent DNA cytidine deaminase encoded by Aicda, and Blimp-1, a transcription factor encoded by Prdm1. B cell-intrinsic expression of Aicda and Prdm1 is regulated by epigenetic elements and processes, including DNA methylation, histone post-translational modifications and non-coding RNAs, particularly miRNAs. Here, we will discuss: B cell-intrinsic epigenetic processes that regulate antibody and autoantibody responses; how epigenetic dysregulation alters CSR/SHM and plasma cell differentiation, thereby leading to autoantibody responses, as in systemic lupus; and, how these can be modulated by nutrients, metabolites, and hormones through changes in B cell-intrinsic epigenetic mechanisms, which can provide therapeutic targets in autoimmunity.


Subject(s)
Antibodies/immunology , Epigenesis, Genetic/immunology , Animals , Antibodies/genetics , Epigenesis, Genetic/genetics , Humans
8.
Nat Commun ; 11(1): 5435, 2020 10 28.
Article in English | MEDLINE | ID: mdl-33116135

ABSTRACT

Memory B cells (MBCs) are long-lived and produce high-affinity, generally, class-switched antibodies. Here, we use a multiparameter approach involving CD27 to segregate naïve B cells (NBC), IgD+ unswitched (unsw)MBCs and IgG+ or IgA+ class-switched (sw)MBCs from humans of different age, sex and race. Conserved antibody variable gene expression indicates that MBCs emerge through unbiased selection from NBCs. Integrative analyses of mRNAs, miRNAs, lncRNAs, chromatin accessibility and cis-regulatory elements uncover a core mRNA-ncRNA transcriptional signature shared by IgG+ and IgA+ swMBCs and distinct from NBCs, while unswMBCs display a transitional transcriptome. Some swMBC transcriptional signature loci are accessible but not expressed in NBCs. Profiling miRNAs reveals downregulated MIR181, and concomitantly upregulated MIR181 target genes such as RASSF6, TOX, TRERF1, TRPV3 and RORα, in swMBCs. Finally, lncRNAs differentially expressed in swMBCs cluster proximal to the IgH chain locus on chromosome 14. Our findings thus provide new insights into MBC transcriptional programs and epigenetic regulation, opening new investigative avenues on these critical cell elements in human health and disease.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory/genetics , Adult , B-Lymphocytes/classification , B-Lymphocytes/metabolism , Chromatin/genetics , Chromatin/immunology , Complementarity Determining Regions , Epigenesis, Genetic , Female , Gene Expression Profiling , Humans , Immunoglobulin Class Switching/genetics , Immunoglobulin Heavy Chains/genetics , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Point Mutation , Signal Transduction/genetics , Transcription Factors/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Young Adult
9.
Sci Adv ; 6(14): eaay2793, 2020 04.
Article in English | MEDLINE | ID: mdl-32270032

ABSTRACT

Activation-induced cytidine deaminase (AID) mediates immunoglobulin class switch DNA recombination (CSR) and somatic hypermutation (SHM), critical processes for maturation of the antibody response. Epigenetic factors, such as histone deacetylases (HDACs), would underpin B cell differentiation stage-specific AID expression. Here, we showed that NAD+-dependent class III HDAC sirtuin 1 (Sirt1) is highly expressed in resting B cells and down-regulated by stimuli inducing AID. B cell Sirt1 down-regulation, deprivation of NAD+ cofactor, or genetic Sirt1 deletion reduced deacetylation of Aicda promoter histones, Dnmt1, and nuclear factor-κB (NF-κB) p65 and increased AID expression. This promoted class-switched and hypermutated T-dependent and T-independent antibody responses or led to generation of autoantibodies. Genetic Sirt1 overexpression, Sirt1 boost by NAD+, or allosteric Sirt1 enhancement by SRT1720 repressed AID expression and CSR/SHM. By deacetylating histone and nonhistone proteins (Dnmt1 and NF-κB p65), Sirt1 transduces metabolic cues into epigenetic changes to play an important B cell-intrinsic role in modulating antibody and autoantibody responses.


Subject(s)
Antibody Formation/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Cytidine Deaminase/genetics , Epigenesis, Genetic , Sirtuin 1/metabolism , Animals , Antibody Formation/immunology , Biomarkers , Cytidine Deaminase/metabolism , DNA Methylation , Humans , Immunophenotyping , Mice , Promoter Regions, Genetic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Front Immunol ; 11: 491, 2020.
Article in English | MEDLINE | ID: mdl-32265934

ABSTRACT

Estrogen contributes to females' strong antibody response to microbial vaccines and proneness to autoimmunity, particularly antibody-mediated systemic autoimmunity, in females. We have hypothesized that this is due to estrogen-mediated potentiation of class switch DNA recombination (CSR) and somatic hypermutation (SHM). As we have shown, estrogen boosts AID expression, which is critical for both CSR and SHM, through upregulation of HoxC4, which together with NF-κB critically mediates Aicda (AID gene) promoter activation. We contend here that additional regulation of Aicda expression by estrogen occurs through epigenetic mechanisms. As we have shown, histone deacetylase inhibitors (HDIs) short-chain fatty acid (SCFA) butyrate and propionate as well as the pharmacologic HDI valproic acid upregulate miRNAs that silence AID expression, thereby modulating specific antibody responses in C57BL/6 mice and autoantibody responses in lupus-prone MRL/Faslpr/lpr mice. Here, using constitutive knockout Esr1-/- mice and B cells as well as conditional knockout Aicdacre/creEsr1flox/flox mice and B cells, we showed that the HDI-mediated downregulation of Aicda expression as well as the maturation of antibody and autoantibody responses is reversed by estrogen and enhanced by deletion of ERα or E2 inhibition. Estrogen's reversion of HDI-mediated inhibition of Aicda and CSR in antibody and autoantibody responses occurred through downregulation of B cell miR-26a, which, as we showed, targets Aicda mRNA 3'UTR. miR-26a was significantly upregulated by HDIs. Accordingly, enforced expression of miR-26a reduced Aicda expression and CSR, while miR-26a-sponges (competitive inhibitors of miR-26a) increased Aicda expression and CSR. Thus, our findings show that estrogen reverses the HDI-mediated downregulation of AID expression and CSR through selective modulation of miR-26a. They also provide mechanistic insights into the immunomodulatory activity of this hormone and a proof-of-principle for using combined ER inhibitor-HDI as a potential therapeutic approach.


Subject(s)
Autoantibodies/biosynthesis , B-Lymphocytes/drug effects , Butyrates/pharmacology , Cytidine Deaminase/biosynthesis , Estradiol/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Immunoglobulin Class Switching/drug effects , Isoantibodies/biosynthesis , MicroRNAs/biosynthesis , Propionates/pharmacology , Valproic Acid/pharmacology , 3' Untranslated Regions , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Base Sequence , Binding, Competitive , Cytidine Deaminase/deficiency , Cytidine Deaminase/genetics , Down-Regulation/drug effects , Estrogen Receptor alpha/deficiency , Female , Gene Expression Regulation/drug effects , Humans , Immunoglobulin Class Switching/genetics , Isoantibodies/immunology , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred MRL lpr , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Phosphoprotein Phosphatases/genetics , Phosphoric Monoester Hydrolases/genetics , Promoter Regions, Genetic , Proof of Concept Study , Recombinant Proteins/metabolism , Sequence Alignment , Sex Characteristics , Transduction, Genetic
12.
J Immunol ; 204(5): 1146-1157, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31932498

ABSTRACT

Upon activation by CD40 or TLR signaling, B lymphocytes activate NF-κB to induce activation-induced cytidine deaminase and, therefore, Ig class switch DNA recombination, as central to the maturation of the Ab and autoantibody responses. In this study, we show that NF-κB activation is boosted by colocalization of engaged immune receptors, such as CD40, with RAB7 small GTPase on mature endosomes, in addition to signals emanating from the receptors localized on the plasma membrane, in mouse B cells. In mature endosomes, RAB7 directly interacts with TRAF6 E3 ubiquitin ligase, which catalyzes K63 polyubiquitination for NF-κB activation. RAB7 overexpression in Cd19+/creRosa26fl-STOP-fl-Rab7 mouse B cells upregulates K63 polyubiquitination activity of TRAF6, enhances NF-κB activation and activation-induced cytidine deaminase induction, and boosts IgG Ab and autoantibody levels. This, together with the extensive intracellular localization of CD40 and the strong correlation of RAB7 expression with NF-κB activation in mouse lupus B cells, shows that RAB7 is an integral component of the B cell NF-κB activation machinery, likely through interaction with TRAF6 for the assembly of "intracellular membrane signalosomes."


Subject(s)
B-Lymphocytes/immunology , Endosomes/immunology , Immunoglobulin Class Switching , NF-kappa B/immunology , TNF Receptor-Associated Factor 6/immunology , Ubiquitination/immunology , rab GTP-Binding Proteins/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , B-Lymphocytes/cytology , Endosomes/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , TNF Receptor-Associated Factor 6/genetics , Ubiquitination/genetics , rab GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
13.
Nat Commun ; 11(1): 60, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31896754

ABSTRACT

Short-chain fatty acids (SCFAs) butyrate and propionate are metabolites from dietary fiber's fermentation by gut microbiota that can affect differentiation or functions of T cells, macrophages and dendritic cells. We show here that at low doses these SCFAs directly impact B cell intrinsic functions to moderately enhance class-switch DNA recombination (CSR), while decreasing at higher doses over a broad physiological range, AID and Blimp1 expression, CSR, somatic hypermutation and plasma cell differentiation. In human and mouse B cells, butyrate and propionate decrease B cell Aicda and Prdm1 by upregulating select miRNAs that target Aicda and Prdm1 mRNA-3'UTRs through inhibition of histone deacetylation (HDAC) of those miRNA host genes. By acting as HDAC inhibitors, not as energy substrates or through GPR-engagement signaling in these B cell-intrinsic processes, these SCFAs impair intestinal and systemic T-dependent and T-independent antibody responses. Their epigenetic impact on B cells extends to inhibition of autoantibody production and autoimmunity in mouse lupus models.


Subject(s)
Antibodies/genetics , Epigenesis, Genetic/drug effects , Fatty Acids, Volatile/pharmacology , Gastrointestinal Microbiome/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Autoantibodies/genetics , Autoantibodies/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Butyrates/pharmacology , Cytidine Deaminase/antagonists & inhibitors , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , Dietary Fiber , Fatty Acids, Volatile/isolation & purification , Fatty Acids, Volatile/pharmacokinetics , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Histone Deacetylase Inhibitors/immunology , Histone Deacetylase Inhibitors/pharmacology , Humans , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Mice, Inbred C57BL , Mice, Mutant Strains , Positive Regulatory Domain I-Binding Factor 1/antagonists & inhibitors , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/immunology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Propionates/pharmacology , Tissue Distribution
14.
J Vis Exp ; (127)2017 09 20.
Article in English | MEDLINE | ID: mdl-28994753

ABSTRACT

Antibody responses are accomplished through several critical B cell-intrinsic processes, including somatic hypermutation (SHM), class-switch DNA recombination (CSR), and plasma cell differentiation. In recent years, epigenetic modifications or factors, such as histone deacetylation and microRNAs (miRNAs), have been shown to interact with B-cell genetic programs to shape antibody responses, while the dysfunction of epigenetic factors has been found to lead to autoantibody responses. Analyzing genome-wide miRNA and mRNA expression in B cells in response to epigenetic modulators is important for understanding the epigenetic regulation of B-cell function and antibody response. Here, we demonstrate a protocol for inducing B cells to undergo CSR and plasma cell differentiation, treating these B cells with histone deacetylase (HDAC) inhibitors (HDIs), and analyzing mRNA and microRNA expression. In this protocol, we directly analyze complementary DNA (cDNA) sequences using next-generation mRNA sequencing (mRNA-seq) and miRNA-seq technologies, mapping of the sequencing reads to the genome, and quantitative reverse transcription (qRT)-PCR. With these approaches, we have defined that, in B cells induced to undergo CSR and plasma cell differentiation, HDI, an epigenetic regulator, selectively modulates miRNA and mRNA expression and alters CSR and plasma cell differentiation.


Subject(s)
B-Lymphocytes/immunology , Genome-Wide Association Study/methods , Histone Deacetylase Inhibitors/pharmacology , MicroRNAs/metabolism , RNA, Messenger/metabolism , Animals , Cell Differentiation/physiology , Humans , Mice , MicroRNAs/genetics , RNA, Messenger/genetics , Recombination, Genetic
15.
Nat Commun ; 8: 14244, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176781

ABSTRACT

Antibody class-switch DNA recombination (CSR) is initiated by AID-introduced DSBs in the switch (S) regions targeted for recombination, as effected by Ku70/Ku86-mediated NHEJ. Ku-deficient B cells, however, undergo (reduced) CSR through an alternative(A)-NHEJ pathway, which introduces microhomologies in S-S junctions. As microhomology-mediated end-joining requires annealing of single-strand DNA ends, we addressed the contribution of single-strand annealing factors HR Rad52 and translesion DNA polymerase θ to CSR. Compared with their Rad52+/+ counterparts, which display normal CSR, Rad52-/- B cells show increased CSR, fewer intra-Sµ region recombinations, no/minimal microhomologies in S-S junctions, decreased c-Myc/IgH translocations and increased Ku70/Ku86 recruitment to S-region DSB ends. Rad52 competes with Ku70/Ku86 for binding to S-region DSB ends. It also facilitates a Ku-independent DSB repair, which favours intra-S region recombination and mediates, particularly in Ku absence, inter-S-S recombination, as emphasized by the significantly greater CSR reduction in Rad52-/- versus Rad52+/+ B cells on Ku86 knockdown.


Subject(s)
DNA End-Joining Repair/immunology , Immunoglobulin Class Switching/genetics , Rad52 DNA Repair and Recombination Protein/metabolism , Recombinational DNA Repair/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/immunology , Cytidine Deaminase/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/immunology , DNA-Directed DNA Polymerase/metabolism , Female , Gene Knockdown Techniques , Humans , Immunoglobulin Class Switching/immunology , Immunoglobulin Switch Region/genetics , Ku Autoantigen/genetics , Ku Autoantigen/immunology , Ku Autoantigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Small Interfering/metabolism , Rad52 DNA Repair and Recombination Protein/genetics , Rad52 DNA Repair and Recombination Protein/immunology , Sulfonamides , DNA Polymerase theta
16.
J Immunol ; 197(10): 3792-3805, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27742832

ABSTRACT

IgG autoantibodies mediate pathology in systemic lupus patients and lupus-prone mice. In this study, we showed that the class-switched IgG autoantibody response in MRL/Faslpr/lpr and C57/Sle1Sle2Sle2 mice was blocked by the CID 1067700 compound, which specifically targeted Ras-related in brain 7 (Rab7), an endosome-localized small GTPase that was upregulated in activated human and mouse lupus B cells, leading to prevention of disease development and extension of lifespan. These were associated with decreased IgG-expressing B cells and plasma cells, but unchanged numbers and functions of myeloid cells and T cells. The Rab7 inhibitor suppressed T cell-dependent and T cell-independent Ab responses, but it did not affect T cell-mediated clearance of Chlamydia infection, consistent with a B cell-specific role of Rab7. Indeed, B cells and plasma cells were inherently sensitive to Rab7 gene knockout or Rab7 activity inhibition in class switching and survival, respectively, whereas proliferation/survival of B cells and generation of plasma cells were not affected. Impairment of NF-κB activation upon Rab7 inhibition, together with the rescue of B cell class switching and plasma cell survival by enforced NF-κB activation, indicated that Rab7 mediates these processes by promoting NF-κB activation, likely through signal transduction on intracellular membrane structures. Thus, a single Rab7-inhibiting small molecule can target two stages of B cell differentiation to dampen the pathogenic autoantibody response in lupus.


Subject(s)
Autoantibodies/immunology , B-Lymphocytes/immunology , Heterocyclic Compounds, 2-Ring/pharmacology , Immunoglobulin Class Switching/drug effects , Lupus Erythematosus, Systemic/immunology , Plasma Cells/physiology , Thiourea/analogs & derivatives , rab GTP-Binding Proteins/antagonists & inhibitors , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/physiology , Cell Proliferation/drug effects , Cell Survival , Chlamydia Infections/immunology , Female , Gene Expression Regulation , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/physiopathology , Lymphocyte Activation , Mice , Mice, Inbred MRL lpr , NF-kappa B/drug effects , NF-kappa B/metabolism , Plasma Cells/drug effects , Plasma Cells/immunology , T-Lymphocytes/immunology , Thiourea/pharmacology , Up-Regulation , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/immunology , rab7 GTP-Binding Proteins
17.
Front Immunol ; 6: 627, 2015.
Article in English | MEDLINE | ID: mdl-26697020

ABSTRACT

As we have suggested, epigenetic factors, such as microRNAs (miRNAs), can interact with genetic programs to regulate B cell functions, thereby informing antibody and autoantibody responses. We have shown that histone deacetylase (HDAC) inhibitors (HDI) inhibit the differentiation events critical to the maturation of the antibody response: class-switch DNA recombination (CSR), somatic hypermutation (SHM), and plasma cell differentiation, by modulating intrinsic B cell mechanisms. HDI repress the expression of AID and Blimp-1, which are critical for CSR/SHM and plasma cell differentiation, respectively, in mouse and human B cells by upregulating selected miRNAs that silenced AICDA/Aicda and PRDM1/Prdm1 mRNAs, as demonstrated by multiple qRT-PCRs (J Immunol 193:5933-5950, 2014). To further define the selectivity of HDI-mediated modulation of miRNA and gene expression, we performed genome-wide miRNA-Seq and mRNA-Seq analysis in B cells stimulated by LPS plus IL-4 and treated with HDI or nil. Consistent with what we have shown using qRT-PCR, these HDI-treated B cells displayed reduced expression of Aicda and Prdm1, and increased expression of miR-155, miR-181b, and miR-361, which target Aicda, and miR-23b, miR-30a, and miR-125b, which target Prdm1. In B cells induced to undergo CSR and plasma cell differentiation, about 23% of over 22,000 mRNAs analyzed were expressed at a significantly high copy number (more than 20 copies/cell). Only 18 (0.36%) of these highly expressed mRNAs, including Aicda, Prdm1, and Xbp1, were downregulated by HDI by 50% or more. Further, only 16 (0.30%) of the highly expressed mRNAs were upregulated (more than twofold) by HDI. The selectivity of HDI-mediated modulation of gene expression was emphasized by unchanged expression of the genes that are involved in regulation, targeting, or DNA repair processes of CSR, as well as unchanged expression of the genes encoding epigenetic regulators and factors that are important for cell signaling or apoptosis. Our findings indicate that, in B cells induced to undergo CSR and plasma cell differentiation, HDI modulate selected miRNAs and mRNAs, possibly as a result of HDACs existing in unique contexts of HDAC/cofactor complexes, as occurring in B lymphocytes, particularly when in an activated state.

18.
Front Immunol ; 6: 631, 2015.
Article in English | MEDLINE | ID: mdl-26697022

ABSTRACT

Epigenetic modifications, such as histone post-translational modifications, DNA methylation, and alteration of gene expression by non-coding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), are heritable changes that are independent from the genomic DNA sequence. These regulate gene activities and, therefore, cellular functions. Epigenetic modifications act in concert with transcription factors and play critical roles in B cell development and differentiation, thereby modulating antibody responses to foreign- and self-antigens. Upon antigen encounter by mature B cells in the periphery, alterations of these lymphocytes epigenetic landscape are induced by the same stimuli that drive the antibody response. Such alterations instruct B cells to undergo immunoglobulin (Ig) class switch DNA recombination (CSR) and somatic hypermutation (SHM), as well as differentiation to memory B cells or long-lived plasma cells for the immune memory. Inducible histone modifications, together with DNA methylation and miRNAs modulate the transcriptome, particularly the expression of activation-induced cytidine deaminase, which is essential for CSR and SHM, and factors central to plasma cell differentiation, such as B lymphocyte-induced maturation protein-1. These inducible B cell-intrinsic epigenetic marks guide the maturation of antibody responses. Combinatorial histone modifications also function as histone codes to target CSR and, possibly, SHM machinery to the Ig loci by recruiting specific adaptors that can stabilize CSR/SHM factors. In addition, lncRNAs, such as recently reported lncRNA-CSR and an lncRNA generated through transcription of the S region that form G-quadruplex structures, are also important for CSR targeting. Epigenetic dysregulation in B cells, including the aberrant expression of non-coding RNAs and alterations of histone modifications and DNA methylation, can result in aberrant antibody responses to foreign antigens, such as those on microbial pathogens, and generation of pathogenic autoantibodies, IgE in allergic reactions, as well as B cell neoplasia. Epigenetic marks would be attractive targets for new therapeutics for autoimmune and allergic diseases, and B cell malignancies.

19.
Cancer Res Treat ; 47(2): 313-21, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25578058

ABSTRACT

PURPOSE: Autophagy is one of the ways to degrade unfolded proteins after endoplasmic reticulum (ER) stress. The purpose of this study is to determine whether a blockade of autophagy leads to aggravated endoplasmic reticulum stress, which then induces cells apoptosis in HeLa cells treated with paclitaxel. MATERIALS AND METHODS: Autophagy activation and the proapoptotic effects were characterized using monodansylcadaverine labeling and Hoechest staining, respectively. A Western blot analysis was used to detect the expression of apoptotic and autophagy-related genes. A flow cytometry was used to assess the cell apoptosis ratio. RESULTS: Paclitaxel exposure induced the aggregation of autophagosomes in the cytoplasms of cervical cancer HeLa cells. The expression of Beclin 1 and LC3 II were upregulated, but p62 was downregulated, which suggests that autophagy was promoted by paclitaxel. On the other hand, the expression of GRP78 obviously increased, suggesting that ER stress was induced after paclitaxel treatment. The cell proliferation assay indicated that a knockdown of Beclin 1 sensitized HeLa cells to paclitaxel. Furthermore, paclitaxel-mediated apoptotic cell death was further potentiated by the pretreatment with autophagy inhibitor chloroquine or small interfering RNA against Beclin 1. These results suggest that an induction of autophagy by paclitaxel may induce cell survival rather than cell death in HeLa cells; moreover, inhibition of autophagy led to an aggravated ER stress and an induction of downstream apoptosis. CONCLUSION: Our results reveal autophagy induced by paclitaxel conferred protection of tumor cells against apoptosis, and blockade of autophagy subsequently aggravated ER stress, enhancing the apoptosis associated with paclitaxel treatment in HeLa cells.

20.
Front Immunol ; 6: 656, 2015.
Article in English | MEDLINE | ID: mdl-26793194
SELECTION OF CITATIONS
SEARCH DETAIL
...