Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Immunol ; 23(7): 1076-1085, 2022 07.
Article in English | MEDLINE | ID: mdl-35761085

ABSTRACT

Memory B cells persist for a lifetime and rapidly differentiate into antibody-producing plasmablasts and plasma cells upon antigen re-encounter. The clonal relationship and evolution of memory B cells and circulating plasmablasts is not well understood. Using single-cell sequencing combined with isolation of specific antibodies, we found that in two healthy donors, the memory B cell repertoire was dominated by large IgM, IgA and IgG2 clonal families, whereas IgG1 families, including those specific for recall antigens, were of small size. Analysis of multiyear samples demonstrated stability of memory B cell clonal families and revealed that a large fraction of recently generated plasmablasts was derived from long-term memory B cell families and was found recurrently. Collectively, this study provides a systematic description of the structure, stability and dynamics of the human memory B cell pool and suggests that memory B cells may be active at any time point in the generation of plasmablasts.


Subject(s)
Memory B Cells , Plasma Cells , B-Lymphocytes , Cells, Cultured , Humans , Immunoglobulin G , Immunologic Memory
2.
Nat Immunol ; 20(10): 1372-1380, 2019 10.
Article in English | MEDLINE | ID: mdl-31451789

ABSTRACT

In multicellular organisms, duplicated genes can diverge through tissue-specific gene expression patterns, as exemplified by highly regulated expression of RUNX transcription factor paralogs with apparent functional redundancy. Here we asked what cell-type-specific biologies might be supported by the selective expression of RUNX paralogs during Langerhans cell and inducible regulatory T cell differentiation. We uncovered functional nonequivalence between RUNX paralogs. Selective expression of native paralogs allowed integration of transcription factor activity with extrinsic signals, while non-native paralogs enforced differentiation even in the absence of exogenous inducers. DNA binding affinity was controlled by divergent amino acids within the otherwise highly conserved RUNT domain and evolutionary reconstruction suggested convergence of RUNT domain residues toward submaximal strength. Hence, the selective expression of gene duplicates in specialized cell types can synergize with the acquisition of functional differences to enable appropriate gene expression, lineage choice and differentiation in the mammalian immune system.


Subject(s)
Core Binding Factor alpha Subunits/genetics , Immune System/physiology , Langerhans Cells/physiology , Organ Specificity/genetics , T-Lymphocytes, Regulatory/physiology , Animals , Cell Differentiation , Cell Lineage , Conserved Sequence , Evolution, Molecular , Gene Duplication , Humans , Mammals , Signal Transduction , Transcriptome
3.
PLoS Genet ; 11(2): e1005020, 2015.
Article in English | MEDLINE | ID: mdl-25714103

ABSTRACT

The development and homeostasis of multicellular organisms relies on gene regulation within individual constituent cells. Gene regulatory circuits that increase the robustness of gene expression frequently incorporate microRNAs as post-transcriptional regulators. Computational approaches, synthetic gene circuits and observations in model organisms predict that the co-regulation of microRNAs and their target mRNAs can reduce cell-to-cell variability in the expression of target genes. However, whether microRNAs directly regulate variability of endogenous gene expression remains to be tested in mammalian cells. Here we use quantitative flow cytometry to show that microRNAs impact on cell-to-cell variability of protein expression in developing mouse thymocytes. We find two distinct mechanisms that control variation in the activation-induced expression of the microRNA target CD69. First, the expression of miR-17 and miR-20a, two members of the miR-17-92 cluster, is co-regulated with the target mRNA Cd69 to form an activation-induced incoherent feed-forward loop. Another microRNA, miR-181a, acts at least in part upstream of the target mRNA Cd69 to modulate cellular responses to activation. The ability of microRNAs to render gene expression more uniform across mammalian cell populations may be important for normal development and for disease.


Subject(s)
Cell Survival/genetics , MicroRNAs/genetics , Protein Biosynthesis/genetics , Thymocytes/metabolism , Animals , Cell Line, Tumor , Flow Cytometry , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Mice , RNA, Messenger/biosynthesis
4.
Blood ; 121(10): 1769-82, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23303821

ABSTRACT

Ikaros family DNA-binding proteins are critical regulators of B-cell development. Because the current knowledge of Ikaros targets in B-cell progenitors is limited, we have identified genes that are bound and regulated by Ikaros in pre-B cells. To elucidate the role of Ikaros in B-cell lineage specification and differentiation, we analyzed the differential expression of Ikaros targets during the progression of multipotent to lymphoid-restricted progenitors, B- and T-cell lineage specification, and progression along the B-cell lineage. Ikaros targets accounted for one-half of all genes up-regulated during B-cell lineage specification in vivo, explaining the essential role of Ikaros in this process. Expression of the Ikaros paralogs Ikzf1 and Ikzf3 increases incrementally during B-cell progenitor differentiation, and, remarkably, inducible Ikaros expression in cycling pre-B cells was sufficient to drive transcriptional changes resembling the differentiation of cycling to resting pre-Bcells in vivo. The data suggest that Ikaros transcription factor dosage drives the progression of progenitors along a predetermined lineage by regulating multiple targets in key pathways, including pre-B­cell receptor signaling, cell cycle progression, and lymphocyte receptor rearrangement.Our approachmay be of general use to map the contribution of transcription factors to cell lineage commitment and differentiation.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Cell Lineage , Genome , Ikaros Transcription Factor/metabolism , Precursor Cells, B-Lymphoid/cytology , Transcription Factors/metabolism , Animals , B-Lymphocytes/metabolism , Binding Sites , Cell Cycle , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation , Ikaros Transcription Factor/genetics , Lymphocyte Activation , Mice , Precursor Cells, B-Lymphoid/metabolism , Signal Transduction , Transcription Factors/genetics
5.
Proc Natl Acad Sci U S A ; 106(2): 516-21, 2009 Jan 13.
Article in English | MEDLINE | ID: mdl-19116266

ABSTRACT

B cell development requires the coordinated rearrangement of Ig heavy (IgH) and light chain loci (IgL). Most mature B cells express a single B cell receptor of unique specificity, and a central question in immunology concerns the mechanisms that prevent the productive rearrangement of >1 IgH and IgL allele per cell. Probabilistic models of allelic exclusion maintain that simultaneous rearrangement of both alleles is rare, because the likelihood of undergoing rearrangement is low for a given Ig allele. Strong support for this idea came from studies in which a GFP marker was inserted into the Igk locus. In this system, the probability of high-level germ-line transcription and subsequent locus rearrangement appeared to be low in pre-B cells. Readdressing the validity of GFP expression as a reporter for the level of germ-line transcription, we found a striking discordance between GFP transcript and protein levels at the pre-B cell stage, which is explained at least in part by the developmentally regulated usage of 2 alternative Igk-J germ-line promoters. These results question the validity of the kappa-GFP system as evidence for probabilistic models of allelic exclusion.


Subject(s)
Alleles , B-Lymphocytes/immunology , Gene Rearrangement , Models, Statistical , Receptors, Antigen, B-Cell/genetics , Animals , B-Lymphocytes/cytology , Bone Marrow Cells , Cells, Cultured , Green Fluorescent Proteins , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Light Chains/genetics , Immunoglobulins/genetics , Mice , Transcription, Genetic
6.
Nat Commun ; 13(1): 2464, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35513363

ABSTRACT

Transmission of epigenetic information between generations occurs in nematodes, flies and plants, mediated by specialised small RNA pathways, modified histones and DNA methylation. Similar processes in mammals can also affect phenotype through intergenerational or trans-generational mechanisms. Here we generate a luciferase knock-in reporter mouse for the imprinted Dlk1 locus to visualise and track epigenetic fidelity across generations. Exposure to high-fat diet in pregnancy provokes sustained re-expression of the normally silent maternal Dlk1 in offspring (loss of imprinting) and increased DNA methylation at the somatic differentially methylated region (sDMR). In the next generation heterogeneous Dlk1 mis-expression is seen exclusively among animals born to F1-exposed females. Oocytes from these females show altered gene and microRNA expression without changes in DNA methylation, and correct imprinting is restored in subsequent generations. Our results illustrate how diet impacts the foetal epigenome, disturbing canonical and non-canonical imprinting mechanisms to modulate the properties of successive generations of offspring.


Subject(s)
Epigenesis, Genetic , Genomic Imprinting , Animals , Biological Variation, Population , DNA Methylation , Diet, High-Fat , Female , Mammals , Mice , Pregnancy
7.
Proc Natl Acad Sci U S A ; 105(22): 7797-802, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18509048

ABSTRACT

Regulatory T (Treg) cells safeguard against autoimmunity and immune pathology. Because determinants of the Treg cell fate are not completely understood, we have delineated signaling events that control the de novo expression of Foxp3 in naive peripheral CD4 T cells and in thymocytes. We report that premature termination of TCR signaling and inibition of phosphatidyl inositol 3-kinase (PI3K) p110alpha, p110delta, protein kinase B (Akt), or mammalian target of rapamycin (mTOR) conferred Foxp3 expression and Treg-like gene expression profiles. Conversely, continued TCR signaling and constitutive PI3K/Akt/mTOR activity antagonised Foxp3 induction. At the chromatin level, di- and trimethylation of lysine 4 of histone H3 (H3K4me2 and -3) near the Foxp3 transcription start site (TSS) and within the 5' untranslated region (UTR) preceded active Foxp3 expression and, like Foxp3 inducibility, was lost upon continued TCR stimulation. These data demonstrate that the PI3K/Akt/mTOR signaling network regulates Foxp3 expression.


Subject(s)
Forkhead Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/metabolism , 5' Untranslated Regions/metabolism , Animals , Forkhead Transcription Factors/genetics , Histones/metabolism , Isoenzymes/metabolism , Methylation , Mice , Mice, Inbred Strains , MicroRNAs/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, Antigen, T-Cell/agonists , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases , Transcription Initiation Site , Transforming Growth Factor beta/metabolism
8.
Nat Commun ; 12(1): 99, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397934

ABSTRACT

CD4 and CD8 mark helper and cytotoxic T cell lineages, respectively, and serve as coreceptors for MHC-restricted TCR recognition. How coreceptor expression is matched with TCR specificity is central to understanding CD4/CD8 lineage choice, but visualising coreceptor gene activity in individual selection intermediates has been technically challenging. It therefore remains unclear whether the sequence of coreceptor gene expression in selection intermediates follows a stereotypic pattern, or is responsive to signaling. Here we use single cell RNA sequencing (scRNA-seq) to classify mouse thymocyte selection intermediates by coreceptor gene expression. In the unperturbed thymus, Cd4+Cd8a- selection intermediates appear before Cd4-Cd8a+ selection intermediates, but the timing of these subsets is flexible according to the strength of TCR signals. Our data show that selection intermediates discriminate MHC class prior to the loss of coreceptor expression and suggest a model where signal strength informs the timing of coreceptor gene activity and ultimately CD4/CD8 lineage choice.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cell Lineage/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Core Binding Factor Alpha 3 Subunit/metabolism , Cytokines/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histocompatibility Antigens/metabolism , Lymphocyte Activation/genetics , Mice, Inbred C57BL , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Transcription Factors/metabolism
9.
Immunology ; 129(4): 610-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20074207

ABSTRACT

As alpha-melanocyte-stimulating hormone (alpha-MSH) is released by immunocompetent cells and has potent immunosuppressive properties, it was determined whether human dendritic cells (DCs) express the receptor for this hormone. Reverse transcription-polymerase chain reaction detected messenger RNA specific for all of the known melanocortin receptors in DCs. Mixed lymphocyte reactions also revealed that treatment with [Nle(4), DPhe(7)]-alpha-MSH (NDP-MSH), a potent alpha-MSH analogue, significantly reduced the ability of DCs to stimulate allogeneic T cells. The expression of various cell surface adhesion, maturation and costimulatory molecules on DCs was also investigated. Although treatment with NDP-MSH did not alter the expression of CD83 and major histocompatibility complex class I and II, the surface expression of CD86 (B7.2), intercellular adhesion molecule (ICAM-1/CD54) and CD1a was reduced. In summary, our data indicate that NDP-MSH inhibits the functional activity of DCs, possibly by down-regulating antigen-presenting and adhesion molecules and that these events may be mediated via the extracellular signal-regulated kinase 1 and 2 pathway.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Receptors, Melanocortin/agonists , alpha-MSH/pharmacology , Antigens, CD1/immunology , Antigens, CD1/metabolism , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Down-Regulation/drug effects , Humans , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Melanocortin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous
10.
Mol Cell Biol ; 24(2): 741-56, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701746

ABSTRACT

The molecular mechanisms governing self-renewal, differentiation, and lineage specification remain unknown. Transcriptional profiling is likely to provide insight into these processes but, as yet, has been confined to "static" molecular profiles of stem and progenitors cells. We now provide a comprehensive, statistically robust, and "dynamic" analysis of multipotent hemopoietic progenitor cells undergoing self-renewal in response to interleukin-3 (IL-3) and multilineage differentiation in response to lineage-affiliated cytokines. Cells undergoing IL-3-dependent proliferative self-renewal displayed striking complexity, including expression of genes associated with different lineage programs, suggesting a highly responsive compartment poised to rapidly execute intrinsically or extrinsically initiated cell fate decisions. A remarkable general feature of early differentiation was a resolution of complexity through the downregulation of gene expression. Although effector genes characteristic of mature cells were upregulated late, coincident with morphological changes, lineage-specific changes in gene expression were observed prior to this, identifying genes which may provide early harbingers of unilineage commitment. Of particular interest were genes that displayed differential behavior irrespective of the lineage elaborated, many of which were rapidly downregulated within 4 to 8 h after exposure to a differentiation cue. These are likely to include genes important in self-renewal, the maintenance of multipotentiality, or the negative regulation of differentiation per se.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Division/drug effects , Cell Division/genetics , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , In Vitro Techniques , Interleukin-3/pharmacology , Mice , Multipotent Stem Cells/drug effects , Oligonucleotide Array Sequence Analysis , Signal Transduction
11.
Methods Mol Biol ; 380: 47-57, 2007.
Article in English | MEDLINE | ID: mdl-17876086

ABSTRACT

Dendritic cells (DC) are key regulators of the immune system. They are capable of stimulating lymphocytes to generate potent cell-mediated and humoral immune responses against pathogens and tumor cells. DC not only activate lymphocytes, but can also educate T cells to tolerate self-antigens, thereby minimizing autoimmune reactions. Another peculiarity of the DC system is the large variety of subsets described, both in the human and in the mouse, according to surface phenotype and organ distribution. Different protocols have been developed to differentiate DC from total mouse bone marrow in vitro. Here, we describe the isolation of a specific DC progenitor population, referred to as preimmunocytes, and document protocols for their differentiation into various DC subsets.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Animals , Bone Marrow Cells/cytology , Dendritic Cells/immunology , Mice
12.
Cell Rep ; 18(5): 1090-1099, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28147266

ABSTRACT

Imprinted genes are regulated according to parental origin and can influence embryonic growth and metabolism and confer disease susceptibility. Here, we designed sensitive allele-specific reporters to non-invasively monitor imprinted Cdkn1c expression in mice and showed that expression was modulated by environmental factors encountered in utero. Acute exposure to chromatin-modifying drugs resulted in de-repression of paternally inherited (silent) Cdkn1c alleles in embryos that was temporary and resolved after birth. In contrast, deprivation of maternal dietary protein in utero provoked permanent de-repression of imprinted Cdkn1c expression that was sustained into adulthood and occurred through a folate-dependent mechanism of DNA methylation loss. Given the function of imprinted genes in regulating behavior and metabolic processes in adults, these results establish imprinting deregulation as a credible mechanism linking early-life adversity to later-life outcomes. Furthermore, Cdkn1c-luciferase mice offer non-invasive tools to identify factors that disrupt epigenetic processes and strategies to limit their long-term impact.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/metabolism , Genomic Imprinting/physiology , Alleles , Animals , Chromatin/physiology , DNA Methylation/physiology , Epigenesis, Genetic/physiology , Mice
14.
J Leukoc Biol ; 74(4): 602-10, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12960259

ABSTRACT

B and T lymphocytes develop through a series of cellular stages, which are defined by recombination status of the immunoglobulin and T cell receptor loci and can be separated by analysis of cell-surface markers. We evaluated how well 26 and 41 samples from five and eight developmental stages of B and T cell development, respectively, could be correctly assigned to their lineage of origin and developmental stage by analysis of the expression of 13,026 genes and expressed sequence tags (ESTs). The RNA expression patterns of eight genes correctly classified all 67 samples as belonging to the B cell or to the T cell lineage. Ninety-two to 100% of B-lineage samples could be correctly assigned to the protein-defined developmental stage by the RNA expression pattern of 29 genes. By contrast, RNA expression patterns of 39 genes were necessary to correctly assign 85-100% of T-lineage samples to the correct developmental stage. The sets of genes used for these classifications contain ESTs as well as known genes that have not previously been associated with lymphocyte development. Graphical display of the classifications shows that B-lineage samples are well separated from T-lineage samples, and samples from the five stages of B cell development are well separated from each other. By contrast, samples from the eight stages of T cell development cannot be separated precisely. We conclude that the protein markers currently widely used for separating stages of B cell development better identify molecularly distinct stages than those used for separating stages of T cell development.


Subject(s)
B-Lymphocytes/physiology , Cell Lineage , Gene Expression Profiling , T-Lymphocytes/physiology , Animals , Biomarkers , Mice , Mice, Inbred C57BL
15.
Immunol Lett ; 82(1-2): 35-40, 2002 Jun 03.
Article in English | MEDLINE | ID: mdl-12008032

ABSTRACT

In mice deficient for the transcription factor Pax-5, B cell development is blocked at the pre-B I cell stage. Like wild type, Pax-5-/- pre-B I cells can be grown long-term in vitro in the presence of stromal cells and IL-7. However, unlike their wild type in vitro grown counterparts, Pax-5-/- pre-B I cells posses an extraordinary developmental plasticity showing hematopoeitic stem cell features such as multipotency and self renewing capacity. Here we review and discuss this in vitro and in vivo plasticity of Pax-5-/- pre-B I cells.


Subject(s)
B-Lymphocytes/immunology , DNA-Binding Proteins/physiology , Hematopoietic Stem Cells/immunology , Transcription Factors/physiology , Animals , Cell Lineage , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , Models, Immunological , PAX5 Transcription Factor , Transcription Factors/genetics
16.
J Exp Med ; 211(11): 2281-95, 2014 Oct 20.
Article in English | MEDLINE | ID: mdl-25311506

ABSTRACT

T cell receptor (TCR) signals can elicit full activation with acquisition of effector functions or a state of anergy. Here, we ask whether microRNAs affect the interpretation of TCR signaling. We find that Dicer-deficient CD4 T cells fail to correctly discriminate between activating and anergy-inducing stimuli and produce IL-2 in the absence of co-stimulation. Excess IL-2 production by Dicer-deficient CD4 T cells was sufficient to override anergy induction in WT T cells and to restore inducible Foxp3 expression in Il2-deficient CD4 T cells. Phosphorylation of Akt on S473 and of S6 ribosomal protein was increased and sustained in Dicer-deficient CD4 T cells, indicating elevated mTOR activity. The mTOR components Mtor and Rictor were posttranscriptionally deregulated, and the microRNAs Let-7 and miR-16 targeted the Mtor and Rictor mRNAs. Remarkably, returning Mtor and Rictor to normal levels by deleting one allele of Mtor and one allele of Rictor was sufficient to reduce Akt S473 phosphorylation and to reduce co-stimulation-independent IL-2 production in Dicer-deficient CD4 T cells. These results show that microRNAs regulate the expression of mTOR components in T cells, and that this regulation is critical for the modulation of mTOR activity. Hence, microRNAs contribute to the discrimination between T cell activation and anergy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Clonal Anergy/genetics , Clonal Anergy/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , MicroRNAs/genetics , TOR Serine-Threonine Kinases/metabolism , Animals , Base Sequence , Binding Sites , Gene Expression Regulation , Humans , Interleukin-2/biosynthesis , Mice , Mice, Transgenic , MicroRNAs/chemistry , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/metabolism , Ribonuclease III/deficiency , Ribonuclease III/genetics , Signal Transduction , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/genetics
17.
Cell Stem Cell ; 6(6): 547-56, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20569692

ABSTRACT

Embryonic stem cells (ESCs) are pluripotent, self-renewing, and have the ability to reprogram differentiated cell types to pluripotency upon cellular fusion. Polycomb-group (PcG) proteins are important for restraining the inappropriate expression of lineage-specifying factors in ESCs. To investigate whether PcG proteins are required for establishing, rather than maintaining, the pluripotent state, we compared the ability of wild-type, PRC1-, and PRC2-depleted ESCs to reprogram human lymphocytes. We show that ESCs lacking either PRC1 or PRC2 are unable to successfully reprogram B cells toward pluripotency. This defect is a direct consequence of the lack of PcG activity because it could be efficiently rescued by reconstituting PRC2 activity in PRC2-deficient ESCs. Surprisingly, the failure of PRC2-deficient ESCs to reprogram somatic cells is functionally dominant, demonstrating a critical requirement for PcG proteins in the chromatin-remodeling events required for the direct conversion of differentiated cells toward pluripotency.


Subject(s)
B-Lymphocytes/metabolism , Embryonic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Induced Pluripotent Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Repressor Proteins/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , B-Lymphocytes/pathology , Cell Fusion , Cell Line, Transformed , Cellular Reprogramming/genetics , Embryonic Stem Cells/pathology , Gene Knockout Techniques , Histone-Lysine N-Methyltransferase/genetics , Humans , Induced Pluripotent Stem Cells/pathology , Mice , Neoplastic Stem Cells/pathology , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Repressor Proteins/genetics , Telomerase/biosynthesis , Telomerase/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
18.
Immunol Lett ; 122(1): 37-43, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-19046990

ABSTRACT

Naive CD4 T cells differentiate into functionally distinct T helper (Th) cells subsets or into regulatory T (Treg) cells in response to the cytokine milieu in which they encounter antigen. A recurring theme in post-thymic CD4 T cell differentiation is the cross-regulation of lineage choice by cytokines and transcription factors that are expressed in alternative lineages. For example, TGFbeta induces the de novo expression of the Treg cell signature transcription factor Foxp3, but iTreg differentiation is blocked by high concentrations of the Th2 cytokine IL4. However, whether IL4 can antagonise Foxp3 induction in more physiological settings remains to be addressed. Here we use a co-culture system to demonstrate that IL4 provided by Th2 cells in vitro is sufficient to block Foxp3 induction in naive CD4 T cells. In addition, we find that Foxp3 induction is efficiently blocked not only by the Th2 transcription factor Gata3, but also by PU.1, which is transiently induced during Th2 differentiation. These data suggest that iTreg differentiation may be affected by the polarity of immune responses.


Subject(s)
GATA3 Transcription Factor/metabolism , Interleukin-4/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Coculture Techniques , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , STAT6 Transcription Factor/deficiency , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Trans-Activators/genetics , Trans-Activators/immunology , Transcriptional Activation/immunology
19.
J Exp Med ; 206(11): 2329-37, 2009 Oct 26.
Article in English | MEDLINE | ID: mdl-19841090

ABSTRACT

Runx proteins are essential for hematopoiesis and play an important role in T cell development by regulating key target genes, such as CD4 and CD8 as well as lymphokine genes, during the specialization of naive CD4 T cells into distinct T helper subsets. In regulatory T (T reg) cells, the signature transcription factor Foxp3 interacts with and modulates the function of several other DNA binding proteins, including Runx family members, at the protein level. We show that Runx proteins also regulate the initiation and the maintenance of Foxp3 gene expression in CD4 T cells. Full-length Runx promoted the de novo expression of Foxp3 during inducible T reg cell differentiation, whereas the isolated dominant-negative Runt DNA binding domain antagonized de novo Foxp3 expression. Foxp3 expression in natural T reg cells remained dependent on Runx proteins and correlated with the binding of Runx/core-binding factor beta to regulatory elements within the Foxp3 locus. Our data show that Runx and Foxp3 are components of a feed-forward loop in which Runx proteins contribute to the expression of Foxp3 and cooperate with Foxp3 proteins to regulate the expression of downstream target genes.


Subject(s)
Core Binding Factor alpha Subunits/metabolism , Forkhead Transcription Factors/metabolism , Animals , Core Binding Factor Alpha 3 Subunit/metabolism , Core Binding Factor alpha Subunits/chemistry , Core Binding Factor beta Subunit/metabolism , Feedback, Physiological , Genes, Dominant , Mice , Protein Structure, Tertiary , T-Lymphocytes, Regulatory/metabolism
20.
Cell Cycle ; 7(15): 2296-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18677111

ABSTRACT

Regulatory T (Treg) cells that express the signature transcription factor Foxp3 safeguard against autoimmunity and immune pathology. Recent studies show that a signaling network with the components phosphatidyl inositol 3 kinase (PI3K), Akt, and the mammalian target of rapamycin (mTOR) regulates the de novo expression of Foxp3 in CD4 T cells. In addition to CD4 T cell differentiation, PI3K/Akt/mTOR signaling also controls T cell migration. Here we review the new data, consider their evolutionary context and discuss their potential implications for immunotherapy.


Subject(s)
Cell Differentiation/drug effects , Growth Inhibitors/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology , Animals , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/physiology , Food , Gene Expression Regulation/physiology , Growth Inhibitors/chemistry , Humans , Immunotherapy/methods , Immunotherapy/trends , Models, Biological , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Kinases/physiology , Receptors, Antigen, T-Cell/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL