Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
J Biol Chem ; : 107581, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39025450

ABSTRACT

Because of their ability to induce lymphocyte apoptosis, glucocorticoids (GC) are widely used to treat hematological malignancies such as lymphomas and multiple myeloma. Their effectiveness is often limited, however, due to the development of glucocorticoid resistance by a variety of molecular mechanisms. Here we performed an unbiased genome-wide CRISPR screen with the human T cell leukemia cell line Jurkat to find previously unidentified genes required for GC-induced apoptosis. One such gene was KMT2D (also known as MLL2 or MLL4), which encodes a histone lysine methyltransferase whose mutations are associated with a variety of cancers, blood malignancies in particular, and are considered markers of poor prognosis. Knockout of KMT2D by CRISPR/Cas9 gene editing in Jurkat and several multiple myeloma cell lines downregulated GR protein expression. Surprisingly, this was not due to a reduction in GR transcripts, but rather to a decrease in the protein's half-life, primarily due to proteasomal degradation. Reconstitution of KMT2D expression restored GR levels. In contrast to the known ability of KMT2D to control gene transcription through covalent histone methylation, KMT2D-mediated upregulation of GR levels did not require its methyltransferase activity. Co-immunoprecipitation and proximity ligation assays found constitutive binding of KMT2D to the GR, which was enhanced in the presence of GC. These observations reveal KMT2D to be essential for stabilization of cellular GR levels, and suggest a possible mechanism by which KMT2D mutations may lead to GC resistance in some malignancies.

2.
Sci Immunol ; 8(89): eadi9066, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37948511

ABSTRACT

How CD4+ lineage gene expression is initiated in differentiating thymocytes remains poorly understood. Here, we show that the paralog transcription factors Zfp281 and Zfp148 control both this process and cytokine expression by T helper cell type 2 (TH2) effector cells. Genetic, single-cell, and spatial transcriptomic analyses showed that these factors promote the intrathymic CD4+ T cell differentiation of class II major histocompatibility complex (MHC II)-restricted thymocytes, including expression of the CD4+ lineage-committing factor Thpok. In peripheral T cells, Zfp281 and Zfp148 promoted chromatin opening at and expression of TH2 cytokine genes but not of the TH2 lineage-determining transcription factor Gata3. We found that Zfp281 interacts with Gata3 and is recruited to Gata3 genomic binding sites at loci encoding Thpok and TH2 cytokines. Thus, Zfp148 and Zfp281 collaborate with Gata3 to promote CD4+ T cell development and TH2 cell responses.


Subject(s)
CD4-Positive T-Lymphocytes , Transcription Factors , Animals , Mice , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Methods Mol Biol ; 2580: 89-114, 2023.
Article in English | MEDLINE | ID: mdl-36374452

ABSTRACT

Recent advances have revolutionized the oldest high-throughput single-cell analytical tool, flow cytometry. Fluorescent analyzers and sorters with up to seven lasers and the potential to detect up to 50 parameters are changing the way flow cytometry is used, but old school practices which are inadequate for new technologies remain alive. This chapter summarizes recent advances, explains the most salient new features and offers a step-by-step guide to develop and successfully execute high-dimensional fluorescent flow cytometry experiments.


Subject(s)
Lasers , Light , Flow Cytometry/methods
4.
Science ; 378(6623): 983-989, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36454826

ABSTRACT

Neurons harbor high levels of single-strand DNA breaks (SSBs) that are targeted to neuronal enhancers, but the source of this endogenous damage remains unclear. Using two systems of postmitotic lineage specification-induced pluripotent stem cell-derived neurons and transdifferentiated macrophages-we show that thymidine DNA glycosylase (TDG)-driven excision of methylcytosines oxidized with ten-eleven translocation enzymes (TET) is a source of SSBs. Although macrophage differentiation favors short-patch base excision repair to fill in single-nucleotide gaps, neurons also frequently use the long-patch subpathway. Disrupting this gap-filling process using anti-neoplastic cytosine analogs triggers a DNA damage response and neuronal cell death, which is dependent on TDG. Thus, TET-mediated active DNA demethylation promotes endogenous DNA damage, a process that normally safeguards cell identity but can also provoke neurotoxicity after anticancer treatments.


Subject(s)
DNA Breaks, Single-Stranded , DNA Demethylation , DNA Repair , Enhancer Elements, Genetic , Induced Pluripotent Stem Cells , Neurons , Thymine DNA Glycosylase , Cell Differentiation , Neurons/enzymology , 5-Methylcytosine/metabolism , Humans , Cell Transdifferentiation
5.
J Extracell Vesicles ; 11(9): e12265, 2022 09.
Article in English | MEDLINE | ID: mdl-36107309

ABSTRACT

CD47 regulates the trafficking of specific coding and noncoding RNAs into extracellular vesicles (EVs), and the RNA contents of CD47+ EVs differ from that of CD63+ EVs released by the same cells. Single particle interferometric reflectance imaging sensing combined with immunofluorescent imaging was used to analyse the colocalization of tetraspanins, integrins, and CD47 on EVs produced by wild type and CD47-deficient Jurkat T lymphoblast and PC3 prostate carcinoma cell lines. On Jurkat cell-derived EVs, ß1 and α4 integrin subunits colocalized predominantly with CD47 and CD81 but not with CD63 and CD9, conserving the known lateral interactions between these proteins in the plasma membrane. Although PC3 cell-derived EVs lacked detectable α4 integrin, specific association of CD81 with ß1 and CD47 was preserved. Loss of CD47 expression in Jurkat cells significantly reduced ß1 and α4 levels on EVs produced by these cells while elevating CD9+ , CD63+ , and CD81+ EVs. In contrast, loss of CD47 in PC3 cells decreased the abundance of CD63+ and CD81+ EVs. These data establish that CD47+ EVs are mostly distinct from EVs bearing the tetraspanins CD63 and CD9, but CD47 also indirectly regulates the abundance of EVs bearing these non-interacting tetraspanins via mechanisms that remain to be determined.


Subject(s)
Carcinoma , Extracellular Vesicles , Prostatic Neoplasms , CD47 Antigen/metabolism , Carcinoma/metabolism , Extracellular Vesicles/metabolism , Humans , Integrin alpha4/metabolism , Integrins/metabolism , Male , Prostate , Prostatic Neoplasms/metabolism , RNA/metabolism , Tetraspanins/metabolism
7.
Nat Med ; 28(7): 1421-1431, 2022 07.
Article in English | MEDLINE | ID: mdl-35501486

ABSTRACT

Despite breakthroughs in cancer immunotherapy, most tumor-reactive T cells cannot persist in solid tumors due to an immunosuppressive environment. We developed Tres (tumor-resilient T cell), a computational model utilizing single-cell transcriptomic data to identify signatures of T cells that are resilient to immunosuppressive signals, such as transforming growth factor-ß1, tumor necrosis factor-related apoptosis-inducing ligand and prostaglandin E2. Tres reliably predicts clinical responses to immunotherapy in melanoma, lung cancer, triple-negative breast cancer and B cell malignancies using bulk T cell transcriptomic data from pre-treatment tumors from patients who received immune-checkpoint inhibitors (n = 38), infusion products for chimeric antigen receptor T cell therapies (n = 34) and pre-manufacture samples for chimeric antigen receptor T cell or tumor-infiltrating lymphocyte therapies (n = 84). Further, Tres identified FIBP, whose functions are largely unknown, as the top negative marker of tumor-resilient T cells across many solid tumor types. FIBP knockouts in murine and human donor CD8+ T cells significantly enhanced T cell-mediated cancer killing in in vitro co-cultures. Further, Fibp knockout in murine T cells potentiated the in vivo efficacy of adoptive cell transfer in the B16 tumor model. Fibp knockout T cells exhibit reduced cholesterol metabolism, which inhibits effector T cell function. These results demonstrate the utility of Tres in identifying biomarkers of T cell effectiveness and potential therapeutic targets for immunotherapies in solid tumors.


Subject(s)
Melanoma , Receptors, Chimeric Antigen , Animals , CD8-Positive T-Lymphocytes , Carrier Proteins , Humans , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Membrane Proteins , Mice
8.
Biomedicines ; 9(11)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34829933

ABSTRACT

T cells and endothelial cells engage in bidirectional communication that regulates angiogenesis and T cell transmigration. Extracellular vesicles (EVs) mediate intercellular communication by the transfer of bioactive molecules including RNAs. EVs produced by a given cell type are heterogeneous in their RNA content, but it is unclear how specific EV surface markers relate to their functional effects on target cells. Our previous work established that Jurkat T cell EVs bearing CD63, MHC-I, or CD47 surface markers contain distinct noncoding RNA populations. The present study reveals that CD63+ and MHC-I+ EVs from CD47-deficient Jurkat T cells are enriched in small non-coding RNAs relative to EVs from wild-type Jurkat T cells. CD47-deficient Jurkat T cells secrete more CD63+ and MHC-I+ EVs, but MHC-I+ EVs are selectively taken up more by human umbilical vein endothelial cells. Transcriptomics analysis of endothelial cells treated with CD63+ or MHC-I+ EVs showed surface marker- and CD47-dependent changes in gene expression in the target cells. Gene set enrichment analysis identified CD47-dependent, and surface marker-dependent effects of T cell EVs on VEGF and inflammatory signaling, cell cycle, and lipid and cholesterol metabolism. Thus, subsets of T cell EVs differentially regulate endothelial cell metabolism and inflammatory and angiogenic responses.

9.
Immunity ; 53(6): 1182-1201.e8, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33242395

ABSTRACT

αß lineage T cells, most of which are CD4+ or CD8+ and recognize MHC I- or MHC II-presented antigens, are essential for immune responses and develop from CD4+CD8+ thymocytes. The absence of in vitro models and the heterogeneity of αß thymocytes have hampered analyses of their intrathymic differentiation. Here, combining single-cell RNA and ATAC (chromatin accessibility) sequencing, we identified mouse and human αß thymocyte developmental trajectories. We demonstrated asymmetric emergence of CD4+ and CD8+ lineages, matched differentiation programs of agonist-signaled cells to their MHC specificity, and identified correspondences between mouse and human transcriptomic and epigenomic patterns. Through computational analysis of single-cell data and binding sites for the CD4+-lineage transcription factor Thpok, we inferred transcriptional networks associated with CD4+- or CD8+-lineage differentiation, and with expression of Thpok or of the CD8+-lineage factor Runx3. Our findings provide insight into the mechanisms of CD4+ and CD8+ T cell differentiation and a foundation for mechanistic investigations of αß T cell development.


Subject(s)
Cell Differentiation/immunology , Cell Lineage/immunology , T-Lymphocyte Subsets/immunology , Thymocytes/immunology , Animals , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Epigenome , Gene Expression Regulation , Gene Regulatory Networks , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens/metabolism , Humans , Mice , T-Lymphocyte Subsets/metabolism , Thymocytes/metabolism , Thymus Gland/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
10.
Proc Natl Acad Sci U S A ; 117(25): 14342-14353, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513716

ABSTRACT

Immature T cells undergo a process of positive selection in the thymus when their new T cell receptor (TCR) engages and signals in response to self-peptides. As the T cell matures, a slew of negative regulatory molecules, including the inhibitory surface glycoprotein CD5, are up-regulated in proportion to the strength of the self-peptide signal. Together these regulators dampen TCR-proximal signaling and help avoid any subsequent peripheral activation of T cells by self-peptides. Paradoxically, antigen-specific T cells initially expressing more CD5 (CD5hi) have been found to better persist as effector/memory cells after a peripheral challenge. The molecular mechanisms underlying such a duality in CD5 function is not clear. We found that CD5 alters the basal activity of the NF-κB signaling in resting peripheral T cells. When CD5 was conditionally ablated, T cells were unable to maintain higher expression of the cytoplasmic NF-κB inhibitor IκBα. Consistent with this, resting CD5hi T cells expressed more of the NF-κB p65 protein than CD5lo cells, without significant increases in transcript levels, in the absence of TCR signals. This posttranslationally stabilized cellular NF-κB depot potentially confers a survival advantage to CD5hi T cells over CD5lo ones. Taken together, these data suggest a two-step model whereby the strength of self-peptide-induced TCR signal lead to the up-regulation of CD5, which subsequently maintains a proportional reserve of NF-κB in peripheral T cells poised for responding to agonistic antigen-driven T cell activation.


Subject(s)
CD5 Antigens/metabolism , Gene Expression Regulation, Developmental/immunology , NF-KappaB Inhibitor alpha/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Antigen Presentation/immunology , CD5 Antigens/genetics , Cell Line, Tumor , Cell Separation , Cell Survival/immunology , Female , Flow Cytometry , Lipopolysaccharides/immunology , Lymphocyte Activation , Mice , Mice, Knockout , Models, Animal , Primary Cell Culture , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology , Transcription Factor RelA/metabolism , Up-Regulation
11.
Cell Rep ; 29(13): 4471-4481.e6, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875554

ABSTRACT

During V(D)J recombination, RAG proteins introduce DNA double-strand breaks (DSBs) at recombination signal sequences (RSSs) that contain either 12- or 23-nt spacer regions. Coordinated 12/23 cleavage predicts that DSBs at variable (V) gene segments should equal the level of breakage at joining (J) segments. Contrary to this, here we report abundant RAG-dependent DSBs at multiple Vκ gene segments independent of V-J rearrangement. We find that a large fraction of Vκ gene segments are flanked not only by a bone-fide 12 spacer but also an overlapping, 23-spacer flipped RSS. These compatible pairs of RSSs mediate recombination and deletion inside the Vκ cluster even in the complete absence of Jκ gene segments and support a V(D)J recombination center (RC) independent of the conventional Jκ-centered RC. We propose an improved model of Vκ-Jκ repertoire formation by incorporating these surprisingly frequent, evolutionarily conserved intra-Vκ cluster recombination events.


Subject(s)
B-Lymphocytes/metabolism , DNA/genetics , Immunoglobulin Variable Region/genetics , V(D)J Recombination/immunology , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , CRISPR-Cas Systems , Clone Cells , DNA/immunology , DNA Breaks, Double-Stranded , DNA Ligase ATP/deficiency , DNA Ligase ATP/genetics , DNA Ligase ATP/immunology , Endonucleases/deficiency , Endonucleases/genetics , Endonucleases/immunology , Female , Gene Editing , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multigene Family , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Spleen/cytology , Spleen/immunology
12.
Nature ; 575(7782): 291-293, 2019 11.
Article in English | MEDLINE | ID: mdl-31719691
13.
PLoS Genet ; 15(4): e1008034, 2019 04.
Article in English | MEDLINE | ID: mdl-31017901

ABSTRACT

Melanocyte stem cells (McSCs) are the undifferentiated melanocytic cells of the mammalian hair follicle (HF) responsible for recurrent generation of a large number of differentiated melanocytes during each HF cycle. HF McSCs reside in both the CD34+ bulge/lower permanent portion (LPP) and the CD34- secondary hair germ (SHG) regions of the HF during telogen. Using Dct-H2BGFP mice, we separate bulge/LPP and SHG McSCs using FACS with GFP and anti-CD34 to show that these two subsets of McSCs are functionally distinct. Genome-wide expression profiling results support the distinct nature of these populations, with CD34- McSCs exhibiting higher expression of melanocyte differentiation genes and with CD34+ McSCs demonstrating a profile more consistent with a neural crest stem cell. In culture and in vivo, CD34- McSCs regenerate pigmentation more efficiently whereas CD34+ McSCs selectively exhibit the ability to myelinate neurons. CD34+ McSCs, and their counterparts in human skin, may be useful for myelinating neurons in vivo, leading to new therapeutic opportunities for demyelinating diseases and traumatic nerve injury.


Subject(s)
Antigens, CD34/metabolism , Melanocytes/immunology , Melanocytes/physiology , Stem Cells/immunology , Stem Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Color/physiology , Hair Follicle/cytology , Hair Follicle/physiology , Melanocytes/classification , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Nude , Mice, Transgenic , Myelin Basic Protein/deficiency , Myelin Basic Protein/genetics , Neural Crest/cytology , Neural Crest/immunology , Neural Crest/physiology , Pigmentation/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regeneration/physiology , Stem Cells/classification
14.
Nat Commun ; 9(1): 5240, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30532006

ABSTRACT

The dynamic nature of the chromatin epigenetic landscape plays a key role in the establishment and maintenance of cell identity, yet the factors that affect the dynamics of the epigenome are not fully known. Here we find that the ubiquitous nucleosome binding proteins HMGN1 and HMGN2 preferentially colocalize with epigenetic marks of active chromatin, and with cell-type specific enhancers. Loss of HMGNs enhances the rate of OSKM induced reprogramming of mouse embryonic fibroblasts (MEFs) into induced pluripotent stem cells (iPSCs), and the ASCL1 induced conversion of fibroblast into neurons. During transcription factor induced reprogramming to pluripotency, loss of HMGNs accelerates the erasure of the MEF-specific epigenetic landscape and the establishment of an iPSCs-specific chromatin landscape, without affecting the pluripotency potential and the differentiation potential of the reprogrammed cells. Thus, HMGN proteins modulate the plasticity of the chromatin epigenetic landscape thereby stabilizing, rather than determining cell identity.


Subject(s)
Cell Membrane/metabolism , Fibroblasts/metabolism , HMGN1 Protein/metabolism , HMGN2 Protein/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Chromatin/genetics , Chromatin/metabolism , Embryo, Mammalian/cytology , Epigenesis, Genetic , Fibroblasts/cytology , HEK293 Cells , HMGN1 Protein/genetics , HMGN2 Protein/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice, Knockout , Mice, Nude , Protein Binding
15.
Hepatology ; 68(1): 127-140, 2018 07.
Article in English | MEDLINE | ID: mdl-29315726

ABSTRACT

Intratumor molecular heterogeneity of hepatocellular carcinoma is partly attributed to the presence of hepatic cancer stem cells (CSCs). Different CSC populations defined by various cell surface markers may contain different oncogenic drivers, posing a challenge in defining molecularly targeted therapeutics. We combined transcriptomic and functional analyses of hepatocellular carcinoma cells at the single-cell level to assess the degree of CSC heterogeneity. We provide evidence that hepatic CSCs at the single-cell level are phenotypically, functionally, and transcriptomically heterogeneous. We found that different CSC subpopulations contain distinct molecular signatures. Interestingly, distinct genes within different CSC subpopulations are independently associated with hepatocellular carcinoma prognosis, suggesting that a diverse hepatic CSC transcriptome affects intratumor heterogeneity and tumor progression. CONCLUSION: Our work provides unique perspectives into the biodiversity of CSC subpopulations, whose molecular heterogeneity further highlights their role in tumor heterogeneity, prognosis, and hepatic CSC therapy. (Hepatology 2018;68:127-140).


Subject(s)
Carcinoma, Hepatocellular/metabolism , Genetic Heterogeneity , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Feasibility Studies , Gene Expression Profiling , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Neoplastic Stem Cells/cytology , Phenotype , Prognosis , Single-Cell Analysis
16.
PLoS One ; 10(11): e0141906, 2015.
Article in English | MEDLINE | ID: mdl-26536348

ABSTRACT

Peripheral T cell lymphoma (PTCL) is a heterogeneous malignancy with poor response to current therapeutic strategies and incompletely characterized genetics. We conducted whole exome sequencing of matched PTCL and non-malignant samples from 12 patients, spanning 8 subtypes, to identify potential oncogenic mutations in PTCL. Analysis of the mutations identified using computational algorithms, CHASM, PolyPhen2, PROVEAN, and MutationAssessor to predict the impact of these mutations on protein function and PTCL tumorigenesis, revealed 104 somatic mutations that were selected as high impact by all four algorithms. Our analysis identified recurrent somatic missense or nonsense mutations in 70 genes, 9 of which contained mutations predicted significant by all 4 algorithms: ATM, RUNX1T1, WDR17, NTRK3, TP53, TRMT12, CACNA2D1, INTS8, and KCNH8. We observed somatic mutations in ATM (ataxia telangiectasia-mutated) in 5 out of the 12 samples and mutations in the common gamma chain (γc) signaling pathway (JAK3, IL2RG, STAT5B) in 3 samples, all of which also harbored mutations in ATM. Our findings contribute insights into the genetics of PTCL and suggest a relationship between γc signaling and ATM in T cell malignancy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Immunoglobulin gamma-Chains/genetics , Lymphoma, T-Cell, Peripheral/genetics , Mutation/genetics , Tumor Suppressor Proteins/genetics , Algorithms , Exome/genetics , Female , Flow Cytometry , Humans , Male
17.
Blood ; 124(25): 3758-67, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25320244

ABSTRACT

Human diffuse large B-cell lymphomas (DLBCLs) often aberrantly express oncogenes that generally contain complex secondary structures in their 5' untranslated region (UTR). Oncogenes with complex 5'UTRs require enhanced eIF4A RNA helicase activity for translation. PDCD4 inhibits eIF4A, and PDCD4 knockout mice have a high penetrance for B-cell lymphomas. Here, we show that on B-cell receptor (BCR)-mediated p70s6K activation, PDCD4 is degraded, and eIF4A activity is greatly enhanced. We identified a subset of genes involved in BCR signaling, including CARD11, BCL10, and MALT1, that have complex 5'UTRs and encode proteins with short half-lives. Expression of these known oncogenic proteins is enhanced on BCR activation and is attenuated by the eIF4A inhibitor Silvestrol. Antigen-experienced immunoglobulin (Ig)G(+) splenic B cells, from which most DLBCLs are derived, have higher levels of eIF4A cap-binding activity and protein translation than IgM(+) B cells. Our results suggest that eIF4A-mediated enhancement of oncogene translation may be a critical component for lymphoma progression, and specific targeting of eIF4A may be an attractive therapeutic approach in the management of human B-cell lymphomas.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Guanylate Cyclase/metabolism , Receptors, Antigen, B-Cell/metabolism , 5' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , B-Cell CLL-Lymphoma 10 Protein , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Blotting, Western , CARD Signaling Adaptor Proteins/genetics , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cells, Cultured , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/genetics , Guanylate Cyclase/genetics , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Middle Aged , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Biosynthesis/drug effects , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Triterpenes/pharmacology
18.
J Immunol ; 192(1): 169-77, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24307734

ABSTRACT

αß T cells, which express the α-ß TCR heterodimer, express CD4 or CD8 coreceptors on cells that are MHC class I or MHC class II dependent. In contrast, γδ T cells do not express CD4 or CD8 and develop independently of MHC interaction. The factors that determine αß and γδ lineage choice are not fully understood, and the determinants of MHC restriction of TCR specificity have been controversial. In this study we have identified a naturally occurring population of T cells expressing Vγ-Cß receptor chains on the cell surface, the products of genomic trans-rearrangement between the Vγ2 gene and a variety of Dß or Jß genes, in place of an intact TCRß-chain and in association with TCRα. Identification of this population allowed an analysis of the role of TCR variable regions in determining T cell lineage choice and MHC restriction. We found that Vγ2(+)Cß(+) cells are positive for either CD4 or CD8 and are selected in an MHC class II- or MHC class I-dependent manner, respectively, thus following the differentiation pathway of αß and not γδ cells and demonstrating that Vß V region sequences are not required for selection of an MHC-restricted repertoire.


Subject(s)
Cell Lineage/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Major Histocompatibility Complex/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression , Genes, MHC Class I , Genes, MHC Class II , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Transcriptional Activation
19.
PLoS One ; 8(4): e62188, 2013.
Article in English | MEDLINE | ID: mdl-23626787

ABSTRACT

Generation and resolution of DNA double-strand breaks is required to assemble antigen-specific receptors from the genes encoding V, D, and J gene segments during recombination. The present report investigates the requirement for ataxia telangiectasia-mutated (ATM) kinase, a component of DNA double-strand break repair, during TCRß recombination and in subsequent TCRß-dependent repertoire generation and thymocyte development. CD4(-)CD8(-) double negative stage 2/3 thymocytes from ATM-deficient mice have both an increased frequency of cells with DNA break foci at TCRß loci and reduced Vß-DJß rearrangement. Sequencing of TCRß complementarity-determining region 3 demonstrates that ATM-deficient CD4(+)CD8(+) double positive thymocytes and peripheral T cells have altered processing of coding ends for both in-frame and out-of-frame TCRß rearrangements, providing the unique demonstration that ATM deficiency alters the expressed TCRß repertoire by a selection-independent mechanism. ATMKO thymi exhibit a partial developmental block in DN cells as they negotiate the ß-selection checkpoint to become double negative stage 4 and CD4(+)CD8(+) thymocytes, resulting in reduced numbers of CD4(+)CD8(+) cells. Importantly, expression of a rearranged TCRß transgene substantially reverses this defect in CD4(+)CD8(+) cells, directly linking a requirement for ATM during endogenous TCRß rearrangement to subsequent TCRß-dependent stages of development. These results demonstrate that ATM plays an important role in TCRß rearrangement, generation of the TCRß CDR3 repertoire, and efficient TCRß-dependent T cell development.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Complementarity Determining Regions/genetics , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Division/genetics , Cell Line , Cell Survival/genetics , DNA Breaks, Double-Stranded , Gene Order , Mice , Mice, Knockout , Thymocytes/metabolism
20.
Nucleic Acids Res ; 41(8): 4535-48, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23470994

ABSTRACT

During the CD4(-)CD8(-) (DN) stage of T-cell development, RAG-dependent DNA breaks and V(D)J recombination occur at three T-cell receptor (TCR) loci: TCRß, TCRγ and TCRδ. During this stage, abnormal trans-rearrangements also take place between TCR loci, occurring at increased frequency in absence of the DNA damage response mediator ataxia telangiectasia mutated (ATM). Here, we use this model of physiologic trans-rearrangement to study factors that predispose to rearrangement and the role of ATM in preventing chromosomal translocations. The frequency of DN thymocytes with DNA damage foci at multiple TCR loci simultaneously is increased 2- to 3-fold in the absence of ATM. However, trans-rearrangement is increased 10 000- to 100 000-fold, indicating that ATM function extends beyond timely resolution of DNA breaks. RAG-mediated synaptic complex formation occurs between recombination signal sequences with unequal 12 and 23 base spacer sequences (12/23 rule). TCR trans-rearrangements violate this rule, as we observed similar frequencies of 12/23 and aberrant 12/12 or 23/23 recombination products. This suggests that trans-rearrangements are not the result of trans-synaptic complex formation, but they are instead because of unstable cis synaptic complexes that form simultaneously at distinct TCR loci. Thus, ATM suppresses trans-rearrangement primarily through stabilization of DNA breaks at TCR loci.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Gene Rearrangement, T-Lymphocyte , Protein Serine-Threonine Kinases/physiology , Thymocytes/immunology , Tumor Suppressor Proteins/physiology , V(D)J Recombination , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , DNA/metabolism , DNA Breaks , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , T-Lymphocytes/immunology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...