Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
STAR Protoc ; 3(2): 101388, 2022 06 17.
Article En | MEDLINE | ID: mdl-35600926

Highly enriched germinal center (GC) B cell populations are essential for studying humoral immunity. Current MACS protocols that isolate untouched GC B cells require GC induction and typically require further FACS purification with direct antibody labeling to achieve sufficiently high purities. We present a MACS protocol with progressive and repeated negative selections that yields highly purified untouched GC B cells from both unimmunized and GC-induced mice and allows further FACS isolation of unlabeled GC B cells from remaining debris by scatter.


B-Lymphocytes , Germinal Center , Animals , Antibodies , Immunity, Humoral , Lymphocyte Count , Mice
2.
Cell Rep ; 36(9): 109624, 2021 08 31.
Article En | MEDLINE | ID: mdl-34469734

B cell tolerance prevents autoimmunity by deleting or deactivating autoreactive B cells that otherwise may cause autoantibody-driven disorders, including systemic lupus erythematosus (lupus). Lupus is characterized by immunoglobulin Gs carrying a double-stranded (ds)-DNA autospecificity derived mainly from somatic hypermutation in the germinal center (GC), pointing to a checkpoint breach of GC B cell tolerance that leads to lupus. However, tolerance mechanisms in the GC remain poorly understood. Here, we show that upregulated sphingomyelin synthase 2 (SMS2) in anti-dsDNA GC B cells induces apoptosis by directly activating protein kinase C δ (PKCδ)'s pro-apoptotic activity. This tolerance mechanism prevents lupus autoimmunity in C57/BL6 mice and can be stimulated pharmacologically to inhibit lupus pathogenesis in lupus-prone NZBWF1 mice. Patients with lupus consistently have substantially reduced SMS2 expression in B cells and to an even greater extent in autoimmune-prone, age-associated B cells, suggesting that patients with lupus have insufficient SMS2-regulated B cell tolerance.


Autoimmunity , B-Lymphocytes/enzymology , Germinal Center/enzymology , Immune Tolerance , Lupus Erythematosus, Systemic/enzymology , Protein Kinase C-delta/metabolism , Transferases (Other Substituted Phosphate Groups)/deficiency , Animals , Apoptosis , Autoimmunity/drug effects , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Enzyme Activators/pharmacology , Female , Genetic Predisposition to Disease , Germinal Center/drug effects , Germinal Center/immunology , Germinal Center/pathology , Immune Tolerance/drug effects , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/prevention & control , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Knockout , Protein Kinase C-delta/genetics , Signal Transduction , Transferases (Other Substituted Phosphate Groups)/genetics , Transferases (Other Substituted Phosphate Groups)/metabolism
3.
J Investig Med ; 65(4): 754-758, 2017 04.
Article En | MEDLINE | ID: mdl-28104820

The use of immune checkpoint inhibitors to treat malignant tumors with microsatellite instability is an emerging new modality. This is based on the observations that these tumors may have a high mutation rate-thus a potential source of tumor-specific neoantigens-and harbor infiltrating cytotoxic T cells in response, suggesting that they may be particularly susceptible to immune checkpoint therapy. PUBMED and ASCO library were systematically reviewed to identify all relevant data that involved the use of immune checkpoint inhibitors in the treatment of cancers with microsatellite instability. The manual search retrieved a total of 3 relevant articles and 1 abstract published between 2015 and 2016. A total of 61 patients with colorectal, 3 with ampullary/cholangiocarcinoma, 2 with endometrial carcinomas, 3 with small bowel cancers, 2 with glioblastoma multiforme, and 1 with bladder cancer with reported efficacy results were reviewed. All the patients had stage IV cancer and were treated with immune checkpoint inhibitors until progression of disease or intolerable side effects emerged. The range of objective response rate was 25-71%. Responses were also durable with progression-free survival at 20 weeks of around 67-78% and to 46% at 1 year. The use of immune checkpoint inhibitors is effective in cancers that express microsatellite instability.


DNA Mismatch Repair , Immune System/pathology , Neoplasms/immunology , Neoplasms/pathology , Humans , Microsatellite Instability , Neoplasms/genetics
4.
Mol Immunol ; 47(6): 1195-206, 2010 Mar.
Article En | MEDLINE | ID: mdl-20116104

PreBCR signaling is critical for B cell development and normally depends on the association of a nascent, component Ig H chain with the surrogate L chain (SLC), which helps ensure that only B cells that synthesize structurally sound antibody can develop. How the invariant and lambda-like SLC vets billions of unique V(H) domains for compatibility with polymorphic kappa and lambda L chains is unclear, because the SLC is composed of not only the Ig domains of VpreB and lambda5, but also the unique regions (URs) that reside at what would be the L chain CDR3. We evaluated the contribution of the Ig and UR domains of lambda5 to H chain screening by evaluating the preBCR-forming capability of lambda5 mutants with a diverse panel of H chains. Using transformed mouse B cells, we demonstrate that the Ig domain of lambda5 was sufficient and its UR dispensable for the rejection of V(H)Q52 and V(H)10 SLC-incompatible H chains. In contrast, the lambda5 UR was necessary to discriminate between SLC-incompatible and -compatible V(H)81X H chains. Substituting the Ig domains of lambda5 with equivalent kappa sequences impaired the SLC's ability to escort all H chains to the surface. Two SLC-incompatible H chains were able to form surface BCRs with two kappa L chains, indicating that the SLC's ability to predict the L chain compatibility of a H chain is not absolute. In sum, lambda5 differentially relies on the lambda-like Ig and UR to probe H chain structure to best accommodate diversity among H chains.


Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Light Chains, Surrogate/chemistry , Immunoglobulin Light Chains, Surrogate/immunology , Alleles , Amino Acid Sequence , Animals , Cell Line , Humans , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/immunology , Immunoglobulin kappa-Chains/immunology , Mice , Molecular Sequence Data , Pre-B Cell Receptors/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
5.
J Immunol ; 181(6): 4098-106, 2008 Sep 15.
Article En | MEDLINE | ID: mdl-18768866

The truncated/V(H)-less mouse H chain Dmu forms precursor B cell receptors with the surrogate L chain complex that promotes allelic exclusion but not other aspects of pre-B cell development, causing most progenitor B cells expressing this H chain to be eliminated at the pre-B cell checkpoint. However, there is evidence that Dmu-lambda1 complexes can be made and are positively selected during fetal life but cannot sustain adult B lymphopoiesis. How surrogate and conventional L chains interpret Dmu's unusual structure and how that affects signaling outcome are unclear. Using nonlymphoid and primary mouse B cells, we show that secretion-competent lambda1 L chains could associate with both full-length H chains and Dmu, whereas secretion-incompetent lambda1 L chains could only do so with full-length H chains. In contrast, Dmu could not form receptors with a panel of kappa L chains irrespective of their secretion properties. This was due to an incompatibility of Dmu with the kappa-joining and constant regions. Finally, the Dmu-lambda1 receptor was less active than the full-length mouse mu-lambda1 receptor in promoting growth under conditions of limiting IL-7. Thus, multiple receptor-dependent mechanisms operating at all stages of B cell development limit the contribution of B cells with Dmu H chain alleles to the repertoire.


B-Lymphocyte Subsets/immunology , Cell Differentiation/immunology , Gene Rearrangement, B-Lymphocyte, Light Chain , Immunoglobulin Heavy Chains/physiology , Immunoglobulin Light Chains, Surrogate/physiology , Immunoglobulin kappa-Chains/physiology , Immunoglobulin lambda-Chains/physiology , Stem Cells/immunology , Alleles , Animals , B-Lymphocyte Subsets/metabolism , Cell Differentiation/genetics , Cell Line , Cell Line, Transformed , Cells, Cultured , Humans , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains, Surrogate/biosynthesis , Immunoglobulin Light Chains, Surrogate/genetics , Immunoglobulin kappa-Chains/biosynthesis , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/biosynthesis , Immunoglobulin lambda-Chains/genetics , Mice , Mice, Knockout , Mutagenesis , Receptors, Antigen, B-Cell/biosynthesis , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/physiology , Stem Cells/metabolism
6.
Nat Immunol ; 7(10): 1082-91, 2006 Oct.
Article En | MEDLINE | ID: mdl-16936731

TFE3 and TFEB are broadly expressed transcription factors related to the transcription factor Mitf. Although they have been linked to cytokine signaling pathways in nonlymphoid cells, their function in T cells is unknown. TFE3-deficient mice are phenotypically normal, whereas TFEB deficiency causes early embryonic death. We now show that combined inactivation of TFE3 and TFEB in T cells resulted in a hyper-immunoglobulin M syndrome due to impaired expression of CD40 ligand by CD4(+) T cells. Native TFE3 and TFEB bound to multiple cognate sites in the promoter of the gene encoding CD40 ligand (Cd40lg), and maximum Cd40lg promoter activity and gene expression required TFE3 or TFEB. Thus, TFE3 and TFEB are direct, physiological and mutually redundant activators of Cd40lg expression in activated CD4(+) T cells critical for T cell-dependent antibody responses.


Antibody Formation/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , CD40 Ligand/genetics , Gene Expression Regulation , Hyper-IgM Immunodeficiency Syndrome/genetics , Thymus Gland/immunology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/agonists , CD40 Ligand/analysis , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics
7.
J Immunol ; 176(11): 6862-72, 2006 Jun 01.
Article En | MEDLINE | ID: mdl-16709846

Signals from the precursor BCR (preBCR) cause proliferation and differentiation of progenitor (pro-) B cells into pre-B cells. Given the very low amounts of surface preBCRs and the demonstrated cell autonomy of preBCR signaling, we examined the possible occurrence of preBCR signal propagation from intracellular membranes such as the endoplasmic reticulum (ER) and the trans-Golgi network (TGN) in transformed and primary pro-B cells. PreBCRs composed of normal Ig mu or truncated Dmu heavy chains (HCs) were redirected to intracellular sites via localization sequences appended to the HC cytoplasmic tail. PreBCR complexes retained in the TGN or shunted from the TGN to lysosomes were as or 50% as active as the corresponding wild-type preBCRs in directing preBCR-dependent events, including CD2 and CD22 expression and proliferation in primary pro-B cells. This occurred despite their low to undetectable surface expression in transformed cells, which otherwise allowed significant surface accumulation of wild-type preBCRs. In contrast, ER-retained preBCRs were inactive. These results suggest that preBCR signaling is remarkably tolerant of dramatic changes in its subcellular distribution within post-ER compartments and support the possibility that the preBCR can activate signaling pathways in the TGN as well as the plasma membrane.


B-Lymphocyte Subsets/immunology , Cell Membrane/immunology , Intracellular Membranes/immunology , Membrane Glycoproteins/physiology , Signal Transduction/immunology , Stem Cells/immunology , Amino Acid Sequence , Animals , B-Lymphocyte Subsets/metabolism , Cell Line , Cell Line, Transformed , Cell Membrane/metabolism , Cells, Cultured , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Membrane Glycoproteins/biosynthesis , Mice , Mice, Knockout , Molecular Sequence Data , Pre-B Cell Receptors , Receptors, Antigen, B-Cell , Stem Cells/metabolism
8.
J Biol Chem ; 280(34): 30225-35, 2005 Aug 26.
Article En | MEDLINE | ID: mdl-15994295

Translocations of the genes encoding the related transcription factors TFE3 and TFEB are almost exclusively associated with a rare juvenile subset of renal cell carcinoma and lead to overexpression of TFE3 or TFEB protein sequences. A better understanding of how deregulated TFE3 and TFEB contribute to the transformation process requires elucidating more of the normal cellular processes in which they participate. Here we identify TFE3 and TFEB as cell type-specific leukemia inhibitory factor-responsive activators of E-cadherin. Overexpression of TFE3 or TFEB in 3T3 cells activated endogenous and reporter E-cadherin expression. Conversely, endogenous TFE3 and/or TFEB was required for endogenous E-cadherin expression in primary mouse embryonic fibroblasts and human embryonic kidney cells. Chromatin precipitation analyses and E-cadherin promoter reporter gene assays revealed that E-cadherin induction by TFE3 or TFEB was primarily or exclusively direct and mitogen-activated protein kinase-dependent in those cell types. In mouse embryonic fibroblasts, TFE3 and TFEB activation of E-cadherin was responsive to leukemia inhibitory factor. In 3T3 cells, TFE3 and TFEB expression also induced expression of Wilms' tumor-1, another E-cadherin activator. In contrast, E-cadherin expression in model mouse and canine renal epithelial cell lines was indifferent to inhibition of endogenous TFE3 and/or TFEB and was reduced by TFE3 or TFEB overexpression. These results reveal new cell type-specific activities of TFE3 and TFEB which may be affected by their mutation.


Cadherins/metabolism , Carcinoma/metabolism , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Interleukin-6/metabolism , Kidney Neoplasms/metabolism , Neoplasm Proteins/physiology , Transcription Factors/physiology , WT1 Proteins/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Blotting, Western , Cell Line , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA, Complementary/metabolism , Dogs , Enzyme Activation , Epithelial Cells , Fibroblasts/metabolism , Genes, Reporter , Genetic Vectors , Humans , Immunoprecipitation , Kidney/metabolism , Leukemia Inhibitory Factor , Luciferases/metabolism , MAP Kinase Signaling System , Mice , Microscopy, Confocal , Mutation , NIH 3T3 Cells , Plasmids/metabolism , Promoter Regions, Genetic , Protein Binding , RNA Interference , Retroviridae/genetics , Retroviridae/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection
9.
J Immunol ; 175(1): 358-66, 2005 Jul 01.
Article En | MEDLINE | ID: mdl-15972669

Signals transduced by precursor-BCRs (pre-BCRs) composed of Ig mu heavy chains (HCs) and the surrogate L chain components lambda5 and VpreB are critical for B cell development. A conserved unique region (UR) of lambda5 was shown to activate pre-BCR complexes in transformed cells and to engage putative ligands, but its contribution to pre-B cell development is not known. It is also not clear why the lambda-like sequences in lambda5 are used to select HCs that will associate mainly with kappa L chains. In this study, we show that, in transformed and primary mouse B cell progenitors, receptors containing full-length HCs and lacking the lambda5UR were expressed at higher surface levels, but exhibited reduced activity compared with normal pre-BCRs in supporting developmental changes that accompany the progenitor to pre-B cell transition in primary cell culture systems and in the bone marrow in vivo. In contrast, deletion of the lambda5UR did not change net signaling output by the Dmu-pre-BCR, a developmentally defective receptor that exhibited impaired activity in the primary cell culture system. Moreover, the lambda-like sequences in lambda5 were more accommodating than kappa in supporting surface expression and signaling by the different HCs. These results show that the lambda5UR is important, although not essential, for surrogate L chain-dependent receptor signaling in primary cells, and furthermore may help allow discrimination of signaling competency between normal and Dmu-pre-BCRs. That the lambda-like portion of lambda5 in the absence of the UR was nondiscriminatory suggests that the lambda5UR focuses pre-BCR-dependent selection on the HC V region.


B-Lymphocytes/immunology , Immunoglobulin Light Chains/chemistry , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Receptors, Antigen, B-Cell/metabolism , Animals , B-Lymphocytes/cytology , Cell Transformation, Viral , Cells, Cultured , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeostasis , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light Chains, Surrogate , In Vitro Techniques , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Protein Structure, Tertiary , Signal Transduction
...