Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
BMC Mol Cell Biol ; 23(1): 10, 2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35189816

ABSTRACT

BACKGROUND: Swi6 acts as a transcription factor in budding yeast, functioning in two different heterodimeric complexes, SBF and MBF, that activate the expression of distinct but overlapping sets of genes. Swi6 undergoes regulated changes in nucleocytoplasmic localization throughout the cell cycle that correlate with changes in gene expression. This study investigates how nucleocytoplasmic transport by multiple transport factors may influence specific Swi6 activities. RESULTS: Here we show that the exportin Crm1 is important for Swi6 nuclear export and activity. Loss of a putative Crm1 NES or inhibition of Crm1 activity results in changes in nucleocytoplasmic Swi6 localization. Alteration of the Crm1 NES in Swi6 results in decreased MBF-mediated gene expression, but does not affect SBF reporter expression, suggesting that export of Swi6 by Crm1 regulates a subset of Swi6 transcription activation activity. Finally, alteration of the putative Crm1 NES in Swi6 results in cells that are larger than wild type, and this increase in cell size is exacerbated by deletion of Msn5. CONCLUSIONS: These data provide evidence that Swi6 has at least two different exportins, Crm1 and Msn5, each of which interacts with a distinct nuclear export signal. We identify a putative nuclear export signal for Crm1 within Swi6, and observe that export by Crm1 or Msn5 independently influences Swi6-regulated expression of a different subset of Swi6-controlled genes. These findings provide new insights into the complex regulation of Swi6 transcription activation activity and the role of nucleocytoplasmic shuttling in regulated gene expression.


Subject(s)
Karyopherins , Saccharomyces cerevisiae Proteins , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Karyopherins/genetics , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcriptional Activation , Exportin 1 Protein
3.
Cell Rep ; 37(6): 109961, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758310

ABSTRACT

Following infection or immunization, memory B cells (MBCs) and long-lived plasma cells provide humoral immunity that can last for decades. Most principles of MBC biology have been determined with hapten-protein carrier models or fluorescent protein immunizations. Here, we examine the temporal dynamics of the germinal center (GC) B cell and MBC response following mouse influenza A virus infection. We find that antiviral B cell responses within the lung-draining mediastinal lymph node (mLN) and the spleen are distinct in regard to duration, enrichment for antigen-binding cells, and class switching dynamics. While splenic GCs dissolve after 6 weeks post-infection, mLN hemagglutinin-specific (HA+) GCs can persist for 22 weeks. Persistent GCs continuously differentiate MBCs, with "peak" and "late" GCs contributing equal numbers of HA+ MBCs to the long-lived compartment. Our findings highlight critical aspects of persistent GC responses and MBC differentiation following respiratory virus infection with direct implications for developing effective vaccination strategies.


Subject(s)
Antibodies, Viral/immunology , Germinal Center/immunology , Immunologic Memory , Influenza A virus/physiology , Memory B Cells/immunology , Orthomyxoviridae Infections/immunology , T-Box Domain Proteins/physiology , Animals , Cell Differentiation , Female , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology
4.
Cell Rep ; 35(12): 109286, 2021 06 22.
Article in English | MEDLINE | ID: mdl-34161770

ABSTRACT

B cell responses are critical for antiviral immunity. However, a comprehensive picture of antigen-specific B cell differentiation, clonal proliferation, and dynamics in different organs after infection is lacking. Here, by combining single-cell RNA and B cell receptor (BCR) sequencing of antigen-specific cells in lymph nodes, spleen, and lungs after influenza infection in mice, we identify several germinal center (GC) B cell subpopulations and organ-specific differences that persist over the course of the response. We discover transcriptional differences between memory cells in lungs and lymphoid organs and organ-restricted clonal expansion. Remarkably, we find significant clonal overlap between GC-derived memory and plasma cells. By combining BCR-mutational analyses with monoclonal antibody (mAb) expression and affinity measurements, we find that memory B cells are highly diverse and can be selected from both low- and high-affinity precursors. By linking antigen recognition with transcriptional programming, clonal proliferation, and differentiation, these finding provide important advances in our understanding of antiviral immunity.


Subject(s)
Antigens, Viral/immunology , B-Lymphocytes/immunology , Gene Expression Profiling , Influenza, Human/genetics , Influenza, Human/immunology , Receptors, Antigen, B-Cell/metabolism , Single-Cell Analysis , Animals , Antibodies, Monoclonal/metabolism , Cell Differentiation/genetics , Cell Proliferation , Clone Cells , Germinal Center/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Memory B Cells/metabolism , Mice, Inbred C57BL , Mutation/genetics , Mutation Rate , Organ Specificity , Plasma Cells/metabolism , RNA/metabolism , Transcription, Genetic
5.
Sci Immunol ; 6(60)2021 06 11.
Article in English | MEDLINE | ID: mdl-34117110

ABSTRACT

Effective antiviral immunity requires generation of T and B lymphocytes expressing the transcription factor T-bet, a regulator of type 1 inflammatory responses. Using T-bet expression as an endogenous marker for cells participating in a type 1 response, we report coordinated interactions of T-bet-expressing T and B lymphocytes on the basis of their dynamic colocalization at the T cell zone and B follicle boundary (T-B boundary) and germinal centers (GCs) during lung influenza infection. We demonstrate that the assembly of this circuit takes place in distinct anatomical niches within the draining lymph node, guided by CXCR3 that enables positioning of TH1 cells at the T-B boundary. The encounter of B and TH1 cells at the T-B boundary enables IFN-γ produced by the latter to induce IgG2c class switching. Within GCs, T-bet+ TFH cells represent a specialized stable sublineage required for GC growth but dispensable for IgG2c class switching. Our studies show that during respiratory viral infection, T-bet-expressing T and B lymphocytes form a circuit assembled in a spatiotemporally controlled manner that acts as a functional unit enabling a robust and coherent humoral response tailored for optimal antiviral immunity.


Subject(s)
B-Lymphocytes/immunology , Immunity, Humoral , Influenza, Human/immunology , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Animals , B-Lymphocytes/metabolism , Cell Communication/immunology , Disease Models, Animal , Female , Germinal Center/cytology , Germinal Center/metabolism , Humans , Immunoglobulin Class Switching , Influenza A virus/immunology , Influenza, Human/pathology , Influenza, Human/virology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice , Mice, Transgenic , Nippostrongylus/immunology , Rats , Receptors, CXCR3/metabolism , Strongylida Infections/immunology , Strongylida Infections/parasitology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/metabolism , Th1 Cells/metabolism
6.
Immunity ; 53(5): 952-970.e11, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33098766

ABSTRACT

Precise targeting of activation-induced cytidine deaminase (AID) to immunoglobulin (Ig) loci promotes antibody class switch recombination (CSR) and somatic hypermutation (SHM), whereas AID targeting of non-Ig loci can generate oncogenic DNA lesions. Here, we examined the contribution of G-quadruplex (G4) nucleic acid structures to AID targeting in vivo. Mice bearing a mutation in Aicda (AIDG133V) that disrupts AID-G4 binding modeled the pathology of hyper-IgM syndrome patients with an orthologous mutation, lacked CSR and SHM, and had broad defects in genome-wide AIDG133V chromatin localization. Genome-wide analyses also revealed that wild-type AID localized to MHCII genes, and AID expression correlated with decreased MHCII expression in germinal center B cells and diffuse large B cell lymphoma. Our findings indicate a crucial role for G4 binding in AID targeting and suggest that AID activity may extend beyond Ig loci to regulate the expression of genes relevant to the physiology and pathology of activated B cells.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , G-Quadruplexes , Hyper-IgM Immunodeficiency Syndrome/etiology , Hyper-IgM Immunodeficiency Syndrome/metabolism , Mutation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Computational Biology/methods , Disease Models, Animal , Disease Susceptibility , Enzyme Activation , Fluorescent Antibody Technique , Gene Expression Profiling , Genome-Wide Association Study , Germinal Center/immunology , Germinal Center/metabolism , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Hyper-IgM Immunodeficiency Syndrome/diagnosis , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunophenotyping , Lymphocyte Activation/genetics , Lymphoma, Large B-Cell, Diffuse/etiology , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic
7.
Cell ; 182(2): 297-316.e27, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32619424

ABSTRACT

The most aggressive B cell lymphomas frequently manifest extranodal distribution and carry somatic mutations in the poorly characterized gene TBL1XR1. Here, we show that TBL1XR1 mutations skew the humoral immune response toward generating abnormal immature memory B cells (MB), while impairing plasma cell differentiation. At the molecular level, TBL1XR1 mutants co-opt SMRT/HDAC3 repressor complexes toward binding the MB cell transcription factor (TF) BACH2 at the expense of the germinal center (GC) TF BCL6, leading to pre-memory transcriptional reprogramming and cell-fate bias. Upon antigen recall, TBL1XR1 mutant MB cells fail to differentiate into plasma cells and instead preferentially reenter new GC reactions, providing evidence for a cyclic reentry lymphomagenesis mechanism. Ultimately, TBL1XR1 alterations lead to a striking extranodal immunoblastic lymphoma phenotype that mimics the human disease. Both human and murine lymphomas feature expanded MB-like cell populations, consistent with a MB-cell origin and delineating an unforeseen pathway for malignant transformation of the immune system.


Subject(s)
Immunologic Memory/physiology , Lymphoma, Large B-Cell, Diffuse/pathology , Nuclear Proteins/genetics , Precursor Cells, B-Lymphoid/immunology , Receptors, Cytoplasmic and Nuclear/genetics , Repressor Proteins/genetics , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Chromatin/chemistry , Chromatin/metabolism , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Histone Deacetylases/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 2/chemistry , Nuclear Receptor Co-Repressor 2/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Transcription, Genetic
8.
Proc Natl Acad Sci U S A ; 116(27): 13474-13479, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31213541

ABSTRACT

A major obstacle to vaccination against antigenically variable viruses is skewing of antibody responses to variable immunodominant epitopes. For influenza virus hemagglutinin (HA), the immunodominance of the variable head impairs responses to the highly conserved stem. Here, we show that head immunodominance depends on the physical attachment of head to stem. Stem immunogenicity is enhanced by immunizing with stem-only constructs or by increasing local HA concentration in the draining lymph node. Surprisingly, coimmunization of full-length HA and stem alters stem-antibody class switching. Our findings delineate strategies for overcoming immunodominance, with important implications for human vaccination.


Subject(s)
Antibodies, Neutralizing/immunology , Epitopes/immunology , Hemagglutinins/immunology , Immunodominant Epitopes/immunology , Influenza A virus/immunology , Influenza Vaccines/immunology , Animals , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Female , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Stem Cells/immunology
9.
Cell Rep ; 27(5): 1472-1486.e5, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31042474

ABSTRACT

The immunoglobulin heavy chain (Igh) locus features a dynamic chromatin landscape to promote class switch recombination (CSR), yet the mechanisms that regulate this landscape remain poorly understood. CHD4, a component of the chromatin remodeling NuRD complex, directly binds H3K9me3, an epigenetic mark present at the Igh locus during CSR. We find that CHD4 is essential for early B cell development but is dispensable for the homeostatic maintenance of mature, naive B cells. However, loss of CHD4 in mature B cells impairs CSR because of suboptimal targeting of AID to the Igh locus. Additionally, we find that CHD4 represses p53 expression to promote B cell proliferation. This work reveals distinct roles for CHD4 in B cell development and CSR and links the H3K9me3 epigenetic mark with AID recruitment to the Igh locus.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , DNA Helicases/genetics , Immunoglobulin Class Switching , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Differentiation , Cells, Cultured , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Genes, Immunoglobulin Heavy Chain , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
J Immunol ; 202(11): 3137-3142, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31028119

ABSTRACT

The DNA damage response protein ATM has long been known to influence class switch recombination in ex vivo-cultured B cells. However, an assessment of B cell-intrinsic requirement of ATM in humoral responses in vivo was confounded by the fact that its germline deletion affects T cell function, and B:T cell interactions are critical for in vivo immune responses. In this study, we demonstrate that B cell-specific deletion of ATM in mice leads to reduction in germinal center (GC) frequency and size in response to immunization. We find that loss of ATM induces apoptosis of GC B cells, likely due to unresolved DNA lesions in cells attempting to undergo class-switch recombination. Accordingly, suboptimal GC responses in ATM-deficient animals are characterized by decreased titers of class-switched Abs and decreased rates of somatic hypermutation. These results unmask the critical B cell-intrinsic role of ATM in maintaining an optimal GC response following immunization.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , B-Lymphocytes/physiology , Germinal Center/physiology , T-Lymphocytes/physiology , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cells, Cultured , DNA Repair/genetics , Immunoglobulin Class Switching , Mice , Mice, Knockout , Receptors, Complement 3d/genetics , Somatic Hypermutation, Immunoglobulin
11.
Mol Cell ; 71(2): 332-342.e8, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30017584

ABSTRACT

The modulator of retrovirus infection (MRI or CYREN) is a 30-kDa protein with a conserved N-terminal Ku-binding motif (KBM) and a C-terminal XLF-like motif (XLM). We show that MRI is intrinsically disordered and interacts with many DNA damage response (DDR) proteins, including the kinases ataxia telangiectasia mutated (ATM) and DNA-PKcs and the classical non-homologous end joining (cNHEJ) factors Ku70, Ku80, XRCC4, XLF, PAXX, and XRCC4. MRI forms large multimeric complexes that depend on its N and C termini and localizes to DNA double-strand breaks (DSBs), where it promotes the retention of DDR factors. Mice deficient in MRI and XLF exhibit embryonic lethality at a stage similar to those deficient in the core cNHEJ factors XRCC4 or DNA ligase IV. Moreover, MRI is required for cNHEJ-mediated DSB repair in XLF-deficient lymphocytes. We propose that MRI is an adaptor that, through multivalent interactions, increases the avidity of DDR factors to DSB-associated chromatin to promote cNHEJ.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , Animals , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA Ligase ATP/genetics , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Ku Autoantigen/genetics , Mice
12.
Elife ; 62017 08 01.
Article in English | MEDLINE | ID: mdl-28760200

ABSTRACT

Heterochromatin formed by the SUV39 histone methyltransferases represses transcription from repetitive DNA sequences and ensures genomic stability. How SUV39 enzymes localize to their target genomic loci remains unclear. Here, we demonstrate that chromatin-associated RNA contributes to the stable association of SUV39H1 with constitutive heterochromatin in human cells. We find that RNA associated with mitotic chromosomes is concentrated at pericentric heterochromatin, and is encoded, in part, by repetitive α-satellite sequences, which are retained in cis at their transcription sites. Purified SUV39H1 directly binds nucleic acids through its chromodomain; and in cells, SUV39H1 associates with α-satellite RNA transcripts. Furthermore, nucleic acid binding mutants destabilize the association of SUV39H1 with chromatin in mitotic and interphase cells - effects that can be recapitulated by RNase treatment or RNA polymerase inhibition - and cause defects in heterochromatin function. Collectively, our findings uncover a previously unrealized function for chromatin-associated RNA in regulating constitutive heterochromatin in human cells.


Subject(s)
Heterochromatin/metabolism , Methyltransferases/metabolism , RNA/metabolism , Repressor Proteins/metabolism , Cell Line , Humans , Protein Binding
13.
Proc Natl Acad Sci U S A ; 114(31): 8354-8359, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28724724

ABSTRACT

DNA double-strand breaks (DSBs) serve as obligatory intermediates for Ig heavy chain (Igh) class switch recombination (CSR). The mechanisms by which DSBs are resolved to promote long-range DNA end-joining while suppressing genomic instability inherently associated with DSBs are yet to be fully elucidated. Here, we use a targeted short-hairpin RNA screen in a B-cell lymphoma line to identify the BRCT-domain protein BRIT1 as an effector of CSR. We show that conditional genetic deletion of BRIT1 in mice leads to a marked increase in unrepaired Igh breaks and a significant reduction in CSR in ex vivo activated splenic B cells. We find that the C-terminal tandem BRCT domains of BRIT1 facilitate its interaction with phosphorylated H2AX and that BRIT1 is recruited to the Igh locus in an activation-induced cytidine deaminase (AID) and H2AX-dependent fashion. Finally, we demonstrate that depletion of another BRCT-domain protein, MDC1, in BRIT1-deleted B cells increases the severity of CSR defect over what is observed upon loss of either protein alone. Our results identify BRIT1 as a factor in CSR and demonstrate that multiple BRCT-domain proteins contribute to optimal resolution of AID-induced DSBs.

14.
Int Immunol ; 29(4): 183-196, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28535205

ABSTRACT

During an immune response, activated B cells may undergo class switch recombination (CSR), a molecular rearrangement that allows B cells to switch from expressing IgM and IgD to a secondary antibody heavy chain isotype such as IgG, IgA or IgE. Secondary antibody isotypes provide the adaptive immune system with distinct effector functions to optimally combat various pathogens. CSR occurs between repetitive DNA elements within the immunoglobulin heavy chain (Igh) locus, termed switch (S) regions and requires the DNA-modifying enzyme activation-induced cytidine deaminase (AID). AID-mediated DNA deamination within S regions initiates the formation of DNA double-strand breaks, which serve as biochemical beacons for downstream DNA repair pathways that coordinate the ligation of DNA breaks. Myriad factors contribute to optimal AID targeting; however, many of these factors also localize to genomic regions outside of the Igh locus. Thus, a current challenge is to explain the specific targeting of AID to the Igh locus. Recent studies have implicated noncoding RNAs in CSR, suggesting a provocative mechanism that incorporates Igh-specific factors to enable precise AID targeting. Here, we chronologically recount the rich history of noncoding RNAs functioning in CSR to provide a comprehensive context for recent and future discoveries. We present a model for the RNA-guided targeting of AID that attempts to integrate historical and recent findings, and highlight potential caveats. Lastly, we discuss testable hypotheses ripe for current experimentation, and explore promising ideas for future investigations.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/metabolism , Immunoglobulin Class Switching , Models, Immunological , RNA, Untranslated/genetics , Animals , Cytidine Deaminase/genetics , Humans , Immunomodulation , Lymphocyte Activation , Mice , Molecular Targeted Therapy , Recombination, Genetic
16.
Nature ; 543(7644): 211-216, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28241136

ABSTRACT

P53-binding protein 1 (53BP1) is a multi-functional double-strand break repair protein that is essential for class switch recombination in B lymphocytes and for sensitizing BRCA1-deficient tumours to poly-ADP-ribose polymerase-1 (PARP) inhibitors. Central to all 53BP1 activities is its recruitment to double-strand breaks via the interaction of the tandem Tudor domain with dimethylated lysine 20 of histone H4 (H4K20me2). Here we identify an uncharacterized protein, Tudor interacting repair regulator (TIRR), that directly binds the tandem Tudor domain and masks its H4K20me2 binding motif. Upon DNA damage, the protein kinase ataxia-telangiectasia mutated (ATM) phosphorylates 53BP1 and recruits RAP1-interacting factor 1 (RIF1) to dissociate the 53BP1-TIRR complex. However, overexpression of TIRR impedes 53BP1 function by blocking its localization to double-strand breaks. Depletion of TIRR destabilizes 53BP1 in the nuclear-soluble fraction and alters the double-strand break-induced protein complex centring 53BP1. These findings identify TIRR as a new factor that influences double-strand break repair using a unique mechanism of masking the histone methyl-lysine binding function of 53BP1.


Subject(s)
Carrier Proteins/metabolism , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Tumor Suppressor p53-Binding Protein 1/antagonists & inhibitors , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Binding Sites , DNA Breaks, Double-Stranded , DNA Repair , Female , Humans , Methylation , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Protein Domains , RNA-Binding Proteins , Telomere-Binding Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1/chemistry
17.
J Exp Med ; 214(1): 197-208, 2017 01.
Article in English | MEDLINE | ID: mdl-28011866

ABSTRACT

Generation of cellular heterogeneity is an essential feature of the adaptive immune system. This is best exemplified during humoral immune response when an expanding B cell clone assumes multiple cell fates, including class-switched B cells, antibody-secreting plasma cells, and memory B cells. Although each cell type is essential for immunity, their generation must be exquisitely controlled because a class-switched B cell cannot revert back to the parent isotype, and a terminally differentiated plasma cell cannot contribute to the memory pool. In this study, we show that an environmental sensor, the aryl hydrocarbon receptor (AhR) is highly induced upon B cell activation and serves a critical role in regulating activation-induced cell fate outcomes. We find that AhR negatively regulates class-switch recombination ex vivo by altering activation-induced cytidine deaminase expression. We further demonstrate that AhR suppresses class switching in vivo after influenza virus infection and immunization with model antigens. In addition, by regulating Blimp-1 expression via Bach2, AhR represses differentiation of B cells into plasmablasts ex vivo and antibody-secreting plasma cells in vivo. These experiments suggest that AhR serves as a molecular rheostat in B cells to brake the effector response, possibly to facilitate optimal recall responses. Thus, AhR might represent a novel molecular target for manipulation of B cell responses during vaccination.


Subject(s)
B-Lymphocytes/physiology , Receptors, Aryl Hydrocarbon/physiology , Animals , Cell Differentiation , Cytidine Deaminase/physiology , Female , Immunoglobulin Class Switching , Influenza A Virus, H1N1 Subtype/immunology , Male , Mice , Mice, Inbred C57BL , Plasma Cells/cytology , Polychlorinated Dibenzodioxins/pharmacology , Positive Regulatory Domain I-Binding Factor 1 , T-Lymphocytes/physiology , Transcription Factors/physiology
18.
Cell ; 166(4): 991-1003, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27477514

ABSTRACT

Small immune complexes cause type III hypersensitivity reactions that frequently result in tissue injury. The responsible mechanisms, however, remain unclear and differ depending on target organs. Here, we identify a kidney-specific anatomical and functional unit, formed by resident macrophages and peritubular capillary endothelial cells, which monitors the transport of proteins and particles ranging from 20 to 700 kDa or 10 to 200 nm into the kidney interstitium. Kidney-resident macrophages detect and scavenge circulating immune complexes "pumped" into the interstitium via trans-endothelial transport and trigger a FcγRIV-dependent inflammatory response and the recruitment of monocytes and neutrophils. In addition, FcγRIV and TLR pathways synergistically "super-activate" kidney macrophages when immune complexes contain a nucleic acid. These data identify a physiological function of tissue-resident kidney macrophages and a basic mechanism by which they initiate the inflammatory response to small immune complexes in the kidney.


Subject(s)
Immune Complex Diseases/immunology , Kidney/cytology , Kidney/immunology , Macrophages/immunology , Animals , Antigen-Antibody Complex , Endothelial Cells , Macrophages/cytology , Mice, Inbred C57BL , Microscopy, Immunoelectron , Monocytes/cytology , Monocytes/immunology , Neutrophils/cytology , Neutrophils/immunology , Receptors, IgG/immunology
19.
Nat Rev Immunol ; 16(3): 164-76, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26898111

ABSTRACT

As B cells engage in the immune response, they express activation-induced cytidine deaminase (AID) to initiate the hypermutation and recombination of immunoglobulin genes, which are crucial processes for the efficient recognition and disposal of pathogens. However, AID must be tightly controlled in B cells to minimize off-target mutations, which can drive chromosomal translocations and the development of B cell malignancies, such as lymphomas. Recent genomic and biochemical analyses have begun to unravel the mechanisms of how AID-mediated deamination is targeted outside immunoglobulin genes. Here, we discuss the transcriptional and topological features that are emerging as key drivers of AID promiscuous activity.


Subject(s)
B-Lymphocytes/immunology , Cytidine Deaminase/immunology , Immunoglobulin Class Switching/immunology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Mutation , Animals , Humans , Somatic Hypermutation, Immunoglobulin/immunology , Translocation, Genetic
20.
Mol Biol Cell ; 22(8): 1375-88, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21346187

ABSTRACT

Nuclear pore complexes (NPCs) provide a gateway for the selective transport of macromolecules across the nuclear envelope (NE). Although we have a solid understanding of NPC composition and structure, we do not have a clear grasp of the mechanism of NPC assembly. Here, we demonstrate specific defects in nucleoporin distribution in strains lacking Heh1p and Heh2p-two conserved members of the LEM (Lap2, emerin, MAN1) family of integral inner nuclear membrane proteins. These effects on nucleoporin localization are likely of functional importance as we have defined specific genetic interaction networks between HEH1 and HEH2, and genes encoding nucleoporins in the membrane, inner, and outer ring complexes of the NPC. Interestingly, expression of a domain of Heh1p that resides in the NE lumen is sufficient to suppress both the nucleoporin mislocalization and growth defects in heh1Δpom34Δ strains. We further demonstrate a specific physical interaction between the Heh1p lumenal domain and the massive cadherin-like lumenal domain of the membrane nucleoporin Pom152p. These findings support a role for Heh1p in the assembly or stability of the NPC, potentially through the formation of a lumenal bridge with Pom152p.


Subject(s)
Membrane Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Biological Transport , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Membrane Proteins/genetics , Nuclear Envelope/chemistry , Nuclear Envelope/metabolism , Nuclear Pore/genetics , Nuclear Pore Complex Proteins/genetics , Nuclear Proteins/genetics , Protein Interaction Mapping , Protein Stability , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Analysis , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...