Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 38(8): 947-953, 2020 08.
Article in English | MEDLINE | ID: mdl-32361713

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has shown promise in hematologic malignancies, but its application to solid tumors has been challenging1-4. Given the unique effector functions of macrophages and their capacity to penetrate tumors5, we genetically engineered human macrophages with CARs to direct their phagocytic activity against tumors. We found that a chimeric adenoviral vector overcame the inherent resistance of primary human macrophages to genetic manipulation and imparted a sustained pro-inflammatory (M1) phenotype. CAR macrophages (CAR-Ms) demonstrated antigen-specific phagocytosis and tumor clearance in vitro. In two solid tumor xenograft mouse models, a single infusion of human CAR-Ms decreased tumor burden and prolonged overall survival. Characterization of CAR-M activity showed that CAR-Ms expressed pro-inflammatory cytokines and chemokines, converted bystander M2 macrophages to M1, upregulated antigen presentation machinery, recruited and presented antigen to T cells and resisted the effects of immunosuppressive cytokines. In humanized mouse models, CAR-Ms were further shown to induce a pro-inflammatory tumor microenvironment and boost anti-tumor T cell activity.


Subject(s)
Immunotherapy, Adoptive , Macrophages/physiology , Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Survival , Humans , Immunotherapy , Lung Neoplasms/therapy , Mice , Microscopy, Video , Neoplasms, Experimental
2.
Stem Cells ; 35(4): 1053-1064, 2017 04.
Article in English | MEDLINE | ID: mdl-28009085

ABSTRACT

The molecular pathways regulating lymphoid priming, fate, and development of multipotent bone marrow hematopoietic stem and progenitor cells (HSPCs) that continuously feed thymic progenitors remain largely unknown. While Notch signal is indispensable for T cell specification and differentiation, the downstream effectors are not well understood. PRL2, a protein tyrosine phosphatase that regulates hematopoietic stem cell proliferation and self-renewal, is highly expressed in murine thymocyte progenitors. Here we demonstrate that protein tyrosine phosphatase PRL2 and receptor tyrosine kinase c-Kit are critical downstream targets and effectors of the canonical Notch/RBPJ pathway in early T cell progenitors. While PRL2 deficiency resulted in moderate defects of thymopoiesis in the steady state, de novo generation of T cells from Prl2 null hematopoietic stem cells was significantly reduced following transplantation. Prl2 null HSPCs also showed impaired T cell differentiation in vitro. We found that Notch/RBPJ signaling upregulated PRL2 as well as c-Kit expression in T cell progenitors. Further, PRL2 sustains Notch-mediated c-Kit expression and enhances stem cell factor/c-Kit signaling in T cell progenitors, promoting effective DN1-DN2 transition. Thus, we have identified a critical role for PRL2 phosphatase in mediating Notch and c-Kit signals in early T cell progenitors. Stem Cells 2017;35:1053-1064.


Subject(s)
Immediate-Early Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/cytology , Animals , Animals, Newborn , Cell Differentiation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice, Inbred C57BL , Models, Biological , Signal Transduction , Thymus Gland/metabolism , Up-Regulation
3.
Proc Natl Acad Sci U S A ; 111(46): E4946-53, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25369933

ABSTRACT

Notch is needed for T-cell development and is a common oncogenic driver in T-cell acute lymphoblastic leukemia. The protooncogene c-Myc (Myc) is a critical target of Notch in normal and malignant pre-T cells, but how Notch regulates Myc is unknown. Here, we identify a distal enhancer located >1 Mb 3' of human and murine Myc that binds Notch transcription complexes and physically interacts with the Myc proximal promoter. The Notch1 binding element in this region activates reporter genes in a Notch-dependent, cell-context-specific fashion that requires a conserved Notch complex binding site. Acute changes in Notch activation produce rapid changes in H3K27 acetylation across the entire enhancer (a region spanning >600 kb) that correlate with Myc expression. This broad Notch-influenced region comprises an enhancer region containing multiple domains, recognizable as discrete H3K27 acetylation peaks. Leukemia cells selected for resistance to Notch inhibitors express Myc despite epigenetic silencing of enhancer domains near the Notch transcription complex binding sites. Notch-independent expression of Myc in resistant cells is highly sensitive to inhibitors of bromodomain containing 4 (Brd4), a change in drug sensitivity that is accompanied by preferential association of the Myc promoter with more 3' enhancer domains that are strongly dependent on Brd4 for function. These findings indicate that altered long-range enhancer activity can mediate resistance to targeted therapies and provide a mechanistic rationale for combined targeting of Notch and Brd4 in leukemia.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Leukemic/genetics , Genes, myc , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/metabolism , Animals , Base Sequence , Cell Cycle Proteins , Cell Line, Tumor , Chromatin Immunoprecipitation , Genes, Reporter , Genome-Wide Association Study , Histones/metabolism , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Models, Molecular , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Promoter Regions, Genetic/genetics , Protein Conformation , Receptor, Notch1/antagonists & inhibitors , Sequence Alignment , Sequence Homology, Nucleic Acid , Transcription Factors/antagonists & inhibitors , Transcription, Genetic
4.
Methods Mol Biol ; 1187: 247-54, 2014.
Article in English | MEDLINE | ID: mdl-25053495

ABSTRACT

Genetic gain- and loss-of-function studies have traditionally been used to study transcriptional networks regulated by the Notch signaling pathway; however these techniques lack the ability to resolve primary and secondary transcriptional events. In contrast, the γ-secretase inhibitor (GSI) washout assay takes advantage of the reversibility of GSI, a pharmacological inhibitor of Notch signaling, along with the ability of cycloheximide to prevent secondary transcriptional effects to identify direct Notch pathway targets. Here we review this technique and the technical considerations for adapting this assay to a cell type of choice.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Receptors, Notch/metabolism , Transcriptional Activation , Animals , Cell Culture Techniques/methods , Humans , Receptors, Notch/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/drug effects
5.
Nat Immunol ; 14(12): 1277-84, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185616

ABSTRACT

Notch signaling induces gene expression of the T cell lineage and discourages alternative fate outcomes. Hematopoietic deficiency in the Notch target Hes1 results in severe T cell lineage defects; however, the underlying mechanism is unknown. We found here that Hes1 constrained myeloid gene-expression programs in T cell progenitor cells, as deletion of the myeloid regulator C/EBP-α restored the development of T cells from Hes1-deficient progenitor cells. Repression of Cebpa by Hes1 required its DNA-binding and Groucho-recruitment domains. Hes1-deficient multipotent progenitor cells showed a developmental bias toward myeloid cells and dendritic cells after Notch signaling, whereas Hes1-deficient lymphoid progenitor cells required additional cytokine signaling for diversion into the myeloid lineage. Our findings establish the importance of constraining developmental programs of the myeloid lineage early in T cell development.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , CCAAT-Enhancer-Binding Protein-alpha/immunology , Homeodomain Proteins/immunology , Receptor, Notch1/immunology , T-Lymphocytes/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Line , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Flow Cytometry , Gene Expression/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myeloid Cells/immunology , Myeloid Cells/metabolism , Protein Binding/immunology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Stem Cells/immunology , Stem Cells/metabolism , T-Lymphocytes/metabolism , Transcription Factor HES-1
6.
Immunity ; 39(1): 148-59, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23890069

ABSTRACT

Two models are proposed to explain Notch function during helper T (Th) cell differentiation. One argues that Notch instructs one Th cell fate over the other, whereas the other posits that Notch function is dictated by cytokines. Here we provide a detailed mechanistic study investigating the role of Notch in orchestrating Th cell differentiation. Notch neither instructed Th cell differentiation nor did cytokines direct Notch activity, but instead, Notch simultaneously regulated the Th1, Th2, and Th17 cell genetic programs independently of cytokine signals. In addition to regulating these programs in both polarized and nonpolarized Th cells, we identified Ifng as a direct Notch target. Notch bound the Ifng CNS-22 enhancer, where it synergized with Tbet at the promoter. Thus, Notch acts as an unbiased amplifier of Th cell differentiation. Our data provide a paradigm for Notch in hematopoiesis, with Notch simultaneously orchestrating multiple lineage programs, rather than restricting alternate outcomes.


Subject(s)
Cytokines/immunology , Receptor, Notch1/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Animals , Base Sequence , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Gene Expression/immunology , Host-Parasite Interactions/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Protein Binding/immunology , Receptor, Notch1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Th1 Cells/metabolism , Th1 Cells/parasitology , Th17 Cells/metabolism , Th2 Cells/metabolism , Trichuris/immunology , Trichuris/physiology
7.
Nat Cell Biol ; 15(5): 502-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23604320

ABSTRACT

Although it is well recognized that haematopoietic stem cells (HSCs) develop from a specialized population of endothelial cells known as haemogenic endothelium, the regulatory pathways that control this transition are not well defined. Here we identify Sox17 as a key regulator of haemogenic endothelial development. Analysis of Sox17-GFP reporter mice revealed that Sox17 is expressed in haemogenic endothelium and emerging HSCs and that it is required for HSC development. Using the mouse embryonic stem cell differentiation model, we show that Sox17 is also expressed in haemogenic endothelium generated in vitro and that it plays a pivotal role in the development and/or expansion of haemogenic endothelium through the Notch signalling pathway. Taken together, these findings position Sox17 as a key regulator of haemogenic endothelial and haematopoietic development.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , HMGB Proteins/metabolism , Hemangioblasts/cytology , SOXF Transcription Factors/metabolism , Animals , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development , Embryonic Stem Cells/metabolism , Female , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HMGB Proteins/genetics , Hemangioblasts/metabolism , Hematopoiesis , Luciferases/metabolism , Male , Mice , Nucleotide Motifs , Promoter Regions, Genetic , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , SOXF Transcription Factors/genetics , Signal Transduction , Transfection
8.
PLoS Genet ; 8(8): e1002855, 2012.
Article in English | MEDLINE | ID: mdl-22916024

ABSTRACT

Oncogenes, which are essential for tumor initiation, development, and maintenance, are valuable targets for cancer therapy. However, it remains a challenge to effectively inhibit oncogene activity by targeting their downstream pathways without causing significant toxicity to normal tissues. Here we show that deletion of mir-181a-1/b-1 expression inhibits the development of Notch1 oncogene-induced T cell acute lymphoblastic leukemia (T-ALL). mir-181a-1/b-1 controls the strength and threshold of Notch activity in tumorigenesis in part by dampening multiple negative feedback regulators downstream of NOTCH and pre-T cell receptor (TCR) signaling pathways. Importantly, although Notch oncogenes utilize normal thymic progenitor cell genetic programs for tumor transformation, comparative analyses of mir-181a-1/b-1 function in normal thymocyte and tumor development demonstrate that mir-181a-1/b-1 can be specifically targeted to inhibit tumor development with little toxicity to normal development. Finally, we demonstrate that mir-181a-1/b-1, but not mir-181a-2b-2 and mir-181-c/d, controls the development of normal thymic T cells and leukemia cells. Together, these results illustrate that NOTCH oncogene activity in tumor development can be selectively inhibited by targeting the molecular networks controlled by mir-181a-1/b-1.


Subject(s)
Gene Expression Regulation, Leukemic , MicroRNAs/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptors, Notch/genetics , Signal Transduction/genetics , T-Lymphocytes/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Humans , Mice , MicroRNAs/metabolism , Molecular Targeted Therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , T-Lymphocytes/pathology , Thymus Gland/metabolism , Thymus Gland/pathology
9.
Nature ; 476(7358): 63-8, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21814277

ABSTRACT

The vertebrate thymus provides an inductive environment for T-cell development. Within the mouse thymus, Notch signals are indispensable for imposing the T-cell fate on multipotential haematopoietic progenitors, but the downstream effectors that impart T-lineage specification and commitment are not well understood. Here we show that a transcription factor, T-cell factor 1 (TCF-1; also known as transcription factor 7, T-cell specific, TCF7), is a critical regulator in T-cell specification. TCF-1 is highly expressed in the earliest thymic progenitors, and its expression is upregulated by Notch signals. Most importantly, when TCF-1 is forcibly expressed in bone marrow (BM) progenitors, it drives the development of T-lineage cells in the absence of T-inductive Notch1 signals. Further characterization of these TCF-1-induced cells revealed expression of many T-lineage genes, including T-cell-specific transcription factors Gata3 and Bcl11b, and components of the T-cell receptor. Our data suggest a model where Notch signals induce TCF-1, and TCF-1 in turn imprints the T-cell fate by upregulating expression of T-cell essential genes.


Subject(s)
Cell Differentiation , Cell Lineage , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Female , Genes, Essential , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Mice, Inbred C57BL , Receptor, Notch1/metabolism , Signal Transduction , T Cell Transcription Factor 1/deficiency , T Cell Transcription Factor 1/genetics , Up-Regulation
10.
Gastroenterology ; 139(6): 2113-23, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20801121

ABSTRACT

BACKGROUND & AIMS: The Notch receptor family regulates cell fate through cell-cell communication. CSL (CBF-1/RBP-jκ, Su(H), Lag-1) drives canonical Notch-mediated gene transcription during cell lineage specification, differentiation, and proliferation in the hematopoietic system, the intestine, the pancreas, and the skin. However, the functional roles of Notch in esophageal squamous epithelial biology are unknown. METHODS: Normal esophageal keratinocytes were stimulated with calcium chloride to induce terminal differentiation. The squamous epithelia were reconstituted in organotypic 3-dimensional culture, a form of human tissue engineering. Notch was inhibited in culture with a γ-secretase inhibitor or dominant negative mastermind-like 1 (DNMAML1). The roles of Notch receptors were evaluated by in vitro gain-of-function and loss-of-function experiments. Additionally, DNMAML1 was targeted to the mouse esophagus by cytokeratin K14 promoter-driven Cre (K14Cre) recombination of Lox-STOP-Lox-DNMAML1. Notch-regulated gene expression was determined by reporter transfection, chromatin immunoprecipitation assays, quantitative reverse-transcription polymerase chain reaction, Western blotting, immunofluorescence, and immunohistochemistry. RESULTS: NOTCH1 (N1) was activated at the onset of squamous differentiation in the esophagus. Intracellular domain of N1 (ICN1) directly activated NOTCH3 (N3) transcription, inducing HES5 and early differentiation markers such as involucrin (IVL) and cytokeratin CK13 in a CSL-dependent fashion. N3 enhanced ICN1 activity and was required for squamous differentiation. Loss of Notch signaling in K14Cre;DNMAML1 mice perturbed esophageal squamous differentiation and resulted in N3 loss and basal cell hyperplasia. CONCLUSIONS: Notch signaling is important for esophageal epithelial homeostasis. In particular, the cross talk of N3 with N1 during differentiation provides novel, mechanistic insights into Notch signaling and squamous epithelial biology.


Subject(s)
Esophagus/cytology , Esophagus/physiology , Receptor, Notch1/genetics , Receptors, Notch/genetics , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Cell Line, Transformed , Humans , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mice, Mutant Strains , Receptor, Notch1/metabolism , Receptor, Notch3 , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription, Genetic/physiology
11.
Semin Immunol ; 22(5): 261-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20630772

ABSTRACT

Notch signaling plays multiple roles in T cell development. Following thymic entry, Notch signals are required to specify the T cell fate from a multipotent hematopoietic progenitor. At subsequent steps in early T cell development, Notch provides important differentiation, survival, proliferation and metabolic signals. This review focuses on the multiple functions of Notch in early T cell development, from T cell specification in the thymus through beta selection.


Subject(s)
Cell Differentiation , Receptors, Notch/immunology , Signal Transduction , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Humans , Protein Binding , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Notch/metabolism , Thymus Gland/metabolism
12.
Genes Dev ; 23(14): 1665-76, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19605688

ABSTRACT

Precise control of the timing and magnitude of Notch signaling is essential for the normal development of many tissues, but the feedback loops that regulate Notch are poorly understood. Developing T cells provide an excellent context to address this issue. Notch1 signals initiate T-cell development and increase in intensity during maturation of early T-cell progenitors (ETP) to the DN3 stage. As DN3 cells undergo beta-selection, during which cells expressing functionally rearranged TCRbeta proliferate and differentiate into CD4(+)CD8(+) progeny, Notch1 signaling is abruptly down-regulated. In this report, we investigate the mechanisms that control Notch1 expression during thymopoiesis. We show that Notch1 and E2A directly regulate Notch1 transcription in pre-beta-selected thymocytes. Following successful beta-selection, pre-TCR signaling rapidly inhibits Notch1 transcription via signals that up-regulate Id3, an E2A inhibitor. Consistent with a regulatory role for Id3 in Notch1 down-regulation, post-beta-selected Id3-deficient thymocytes maintain Notch1 transcription, whereas enforced Id3 expression decreases Notch1 expression and abrogates Notch1-dependent T-cell survival. These data provide new insights into Notch1 regulation in T-cell progenitors and reveal a direct link between pre-TCR signaling and Notch1 expression during thymocyte development. Our findings also suggest new strategies for inhibiting Notch1 signaling in pathologic conditions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Genes, T-Cell Receptor beta/physiology , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cell Line , Down-Regulation , Genes, T-Cell Receptor beta/genetics , Homeodomain Proteins/metabolism , Inhibitor of Differentiation Proteins/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/metabolism , Receptor, Notch1/genetics
13.
Cancer Res ; 69(13): 5312-20, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19549918

ABSTRACT

The importance of mitogen-activated protein kinase signaling in melanoma is underscored by the prevalence of activating mutations in N-Ras and B-Raf, yet clinical development of inhibitors of this pathway has been largely ineffective, suggesting that alternative oncogenes may also promote melanoma. Notch is an interesting candidate that has only been correlated with melanoma development and progression; a thorough assessment of tumor-initiating effects of activated Notch on human melanocytes would clarify the mounting correlative evidence and perhaps identify a novel target for an otherwise untreatable disease. Analysis of a substantial panel of cell lines and patient lesions showed that Notch activity is significantly higher in melanomas than their nontransformed counterparts. The use of a constitutively active, truncated Notch transgene construct (N(IC)) was exploited to determine if Notch activation is a "driving" event in melanocytic transformation or instead a "passenger" event associated with melanoma progression. N(IC)-infected melanocytes displayed increased proliferative capacity and biological features more reminiscent of melanoma, such as dysregulated cell adhesion and migration. Gene expression analyses supported these observations and aided in the identification of MCAM, an adhesion molecule associated with acquisition of the malignant phenotype, as a direct target of Notch transactivation. N(IC)-positive melanocytes grew at clonal density, proliferated in limiting media conditions, and also exhibited anchorage-independent growth, suggesting that Notch alone is a transforming oncogene in human melanocytes, a phenomenon not previously described for any melanoma oncogene. This new information yields valuable insight into the basic epidemiology of melanoma and launches a realm of possibilities for drug intervention in this deadly disease.


Subject(s)
Melanocytes/physiology , Receptor, Notch1/physiology , CD146 Antigen/genetics , CD146 Antigen/physiology , Cell Division , Disease Progression , Foreskin/cytology , Humans , Male , Melanocytes/pathology , Melanoma/genetics , Melanoma/pathology , Melanoma/prevention & control , Phenotype , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control , Up-Regulation
14.
Blood ; 110(10): 3706-14, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17671232

ABSTRACT

Gene expression profiling of acute myeloid leukemia (AML) allows the discovery of previously unrecognized molecular entities. Here, we identified a specific subgroup of AML, defined by an expression profile resembling that of AMLs with mutations in the myeloid transcription factor CCAAT/enhancer-binding protein alpha (C/EBPalpha), while lacking such mutations. We found that in these leukemias, the CEBPA gene was silenced, which was associated with frequent promoter hypermethylation. The leukemias phenotypically showed aberrant expression of T-cell genes, of which CD7 was most consistent. We identified 2 mechanisms that may contribute to this phenotype. First, absence of Cebpa led to up-regulation of specific T-cell transcripts (ie, Cd7 and Lck) in hematopoietic stem cells isolated from conditional Cebpa knockout mice. Second, the enhanced expression of TRIB2, which we identify here as a direct target of the T-cell commitment factor NOTCH1, suggested aberrantly activated Notch signaling. Putatively activating NOTCH1 mutations were found in several specimens of the newly identified subgroup, while a large set of control AMLs was mutation negative. A gene expression prediction signature allowed the detection of similar cases of leukemia in independent series of AML.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Gene Silencing , Leukemia, Myeloid, Acute/genetics , Leukemia-Lymphoma, Adult T-Cell/genetics , Receptor, Notch1/genetics , Animals , Cells, Cultured , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , Mice , Mice, Transgenic , Mutation , Oligonucleotide Array Sequence Analysis
15.
J Immunol ; 179(3): 1760-7, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641042

ABSTRACT

OX40 is a member of the TNFR superfamily (CD134; TNFRSF4) that is expressed on activated T cells and regulates T cell-mediated immune responses. In this study, we have examined the regulation of OX40 gene expression in T cells. Low-level OX40 mRNA expression was detected in both resting T cells and the nonactivated EL4 T cell line, and was up-regulated in both types of T cells upon activation with anti-CD3 Ab. We have shown in this study that basal OX40 promoter activity is regulated by constitutively expressed Sp1/Sp3 and YY1 transcription factors. NF-kappaB (p50 and p65) also binds to the OX40 promoter region, but the level of direct enhancement of the OX40 promoter activity by this transcription factor is not sufficient to account for the observed up-regulation of OX40 mRNA expression associated with activation. We have detected by chromatin immunoprecipitation that histone H4 molecules in the OX40 promoter region are highly acetylated by activation and NF-kappaB binds to the OX40 promoter in vivo. These findings suggest that OX40 gene expression is regulated by chromatin remodeling, and that NF-kappaB might be involved in initiation of chromatin remodeling in the OX40 promoter region in activated T cells. CD4(+)CD25(+) regulatory T (Treg) cells also express OX40 at high levels, and signaling through this receptor can neutralize suppressive activity of this Treg cell. In CD4(+)CD25(+) Treg cells, histone H4 molecules in the OX40 promoter region are also highly acetylated, even in the absence of in vitro activation.


Subject(s)
Chromatin Assembly and Disassembly/immunology , NF-kappa B p50 Subunit/metabolism , Promoter Regions, Genetic/immunology , Receptors, OX40/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Up-Regulation/immunology , YY1 Transcription Factor/metabolism , Animals , Binding Sites/genetics , Binding Sites/immunology , CD3 Complex/immunology , Cell Line, Tumor , Chromatin Assembly and Disassembly/genetics , Immune Sera/pharmacology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/metabolism , Mice , RNA, Messenger/biosynthesis , Receptors, OX40/biosynthesis , Receptors, OX40/metabolism , Sp1 Transcription Factor/physiology , Sp3 Transcription Factor/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Factor RelA/metabolism , Up-Regulation/genetics , YY1 Transcription Factor/physiology
16.
Immunity ; 27(1): 100-10, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17658278

ABSTRACT

Notch signaling plays multiple roles to direct diverse decisions regarding cell fate during T cell development. During helper T (Th) cell differentiation, Notch is involved in generating optimal Th2 cell responses. Here, we present data investigating how Notch mediates Th2 cell differentiation. Notch showed a CD4(+) T cell intrinsic role in promoting IL-4 expression that required GATA-3. In the absence of Notch signals, Gata3 expression was markedly diminished. Introduction of an activated allele of Notch1 into CD4(+) T cells led to the specific and direct upregulation of a developmentally regulated Gata3 transcript that included the exon 1a sequences. Furthermore, Notch acted in parallel with GATA-3 to synergistically activate IL-4 expression. Together, these data implicate Gata3 as a direct transcriptional Notch target that acts in concert with Notch signaling to generate optimal Th2 cell responses.


Subject(s)
Cell Differentiation/immunology , GATA3 Transcription Factor/metabolism , Receptors, Notch/physiology , Th2 Cells/cytology , Th2 Cells/metabolism , Animals , Cells, Cultured , Down-Regulation/immunology , GATA3 Transcription Factor/antagonists & inhibitors , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/genetics , Gene Expression Regulation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Signal Transduction/immunology , Th2 Cells/immunology
17.
Blood ; 110(7): 2650-8, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17585052

ABSTRACT

Oncogenic Notch1 mutations are found in most T-lineage acute lymphoblastic leukemias in humans and T-cell lymphomas in mice. However, the mechanism by which Notch1 promotes transformation or maintains malignant cell survival has not been determined fully. Here, we report that expression of the transcription factor lymphoid enhancer factor 1 (Lef1) is Notch dependent in murine T-cell lymphomas in vitro and in vivo, and that the intracellular domain of Notch1 (ICN1) is present at the Lef1 promoter. Lef1 expression is not Notch dependent in primary T-cell progenitors, but Lef1 mRNA is increased by ectopic expression of ICN1 in these cells. We show that Lef1 is required for survival of T-cell lymphoma lines, and that ectopic expression of Lef1 delays lymphoma cell death in the absence of Notch signaling, indicating that Lef1 is an important Notch target in these cells. Therefore, Notch1 co-opts Lef1 during the process of transformation to maintain survival of T-cell lymphomas.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/metabolism , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Receptor, Notch1/metabolism , Animals , Base Sequence , Binding Sites , Cell Survival , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoma, T-Cell/genetics , Mice , Mice, Knockout , Sequence Alignment , Signal Transduction , Stem Cells/metabolism
18.
J Exp Med ; 203(10): 2239-45, 2006 Oct 02.
Article in English | MEDLINE | ID: mdl-16966428

ABSTRACT

Genetic inactivation of Notch signaling in CD4(-)CD8(-) double-negative (DN) thymocytes was previously shown to impair T cell receptor (TCR) gene rearrangement and to cause a partial block in CD4(+)CD8(+) double-positive (DP) thymocyte development in mice. In contrast, in vitro cultures suggested that Notch was absolutely required for the generation of DP thymocytes independent of pre-TCR expression and activity. To resolve the respective role of Notch and the pre-TCR, we inhibited Notch-mediated transcriptional activation in vivo with a green fluorescent protein-tagged dominant-negative Mastermind-like 1 (DNMAML) that allowed us to track single cells incapable of Notch signaling. DNMAML expression in DN cells led to decreased production of DP thymocytes but only to a modest decrease in intracellular TCRbeta expression. DNMAML attenuated the pre-TCR-associated increase in cell size and CD27 expression. TCRbeta or TCRalphabeta transgenes failed to rescue DNMAML-related defects. Intrathymic injections of DNMAML(-) or DNMAML(+) DN thymocytes revealed a complete DN/DP transition block, with production of DNMAML(+) DP thymocytes only from cells undergoing late Notch inactivation. These findings indicate that the Notch requirement during the beta-selection checkpoint in vivo is absolute and independent of the pre-TCR, and it depends on transcriptional activation by Notch via the CSL/RBP-J-MAML complex.


Subject(s)
Cell Differentiation/immunology , Gene Rearrangement/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Notch/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Chromatin Immunoprecipitation , DNA Primers , Flow Cytometry , Green Fluorescent Proteins , Mice , Nuclear Proteins/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Transcription Factors/metabolism , Transcriptional Activation/immunology
19.
Genes Dev ; 20(15): 2096-109, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16847353

ABSTRACT

Human acute T-cell lymphoblastic leukemias and lymphomas (T-ALL) are commonly associated with gain-of-function mutations in Notch1 that contribute to T-ALL induction and maintenance. Starting from an expression-profiling screen, we identified c-myc as a direct target of Notch1 in Notch-dependent T-ALL cell lines, in which Notch accounts for the majority of c-myc expression. In functional assays, inhibitors of c-myc interfere with the progrowth effects of activated Notch1, and enforced expression of c-myc rescues multiple Notch1-dependent T-ALL cell lines from Notch withdrawal. The existence of a Notch1-c-myc signaling axis was bolstered further by experiments using c-myc-dependent murine T-ALL cells, which are rescued from withdrawal of c-myc by retroviral transduction of activated Notch1. This Notch1-mediated rescue is associated with the up-regulation of endogenous murine c-myc and its downstream transcriptional targets, and the acquisition of sensitivity to Notch pathway inhibitors. Additionally, we show that primary murine thymocytes at the DN3 stage of development depend on ligand-induced Notch signaling to maintain c-myc expression. Together, these data implicate c-myc as a developmentally regulated direct downstream target of Notch1 that contributes to the growth of T-ALL cells.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cells, Cultured , Humans , Mice , Proto-Oncogene Proteins c-myc/genetics , Receptor, Notch1/genetics , Thymus Gland/cytology , Thymus Gland/metabolism
20.
J Leukoc Biol ; 78(3): 753-61, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16126843

ABSTRACT

T helper cell type 1 (Th1) and Th2 cells express distinct sets of chemokine receptors. In contrast to Th1 chemokine receptors, it is largely unknown how Th2 chemokine receptors such as CC chemokine receptor 4 (CCR4) are induced during Th2 differentiation. Here, we investigated the induction of CCR4 surface expression and ligand responsiveness evaluated by functional assays such as chemokine binding and chemotaxis. This was done in comparison with those of a Th1 chemokine receptor, CXC chemokine receptor 3 (CXCR3). Resting T cells expressed neither CXCR3 nor CCR4. CXCR3 expression and ligand responsiveness were observed when resting T cells were stimulated with anti-CD3 plus anti-CD28 in the presence of [interleukin (IL)-12+anti-IL-4] and then recultured without T cell receptor (TCR) stimulation. Unlike CXCR3, CCR4 was induced immediately after anti-CD3/anti-CD28 stimulation in the presence of (IL-4+anti-interferon-gamma+anti-IL-12). However, these CCR4-positive cells failed to exhibit chemokine binding and chemotaxis. Although the levels of surface CCR4 expression were not increased after the subsequent reculture in the absence of TCR stimulation, CCR4 responsiveness was induced in this stage of Th2 cells. The induction of CCR4 expression and the acquisition of CCR4 responsiveness did not occur in IL-4-deficient (IL-4(-/-)) and signal transducer and activator of transcription (STAT)6(-/-) T cells. CCR4 expression and functionality were regained in IL-4(-/-) but not in STAT6(-/-) T cells by the addition of recombinant IL-4. Although surface expression and functionality of CCR4 are induced depending on the IL-4/STAT6 signaling pathway, the present results indicate that the functionality of CCR4 does not correlate with CCR4 expression but emerges at later stages of Th2 differentiation.


Subject(s)
Gene Expression Regulation/immunology , Receptors, Chemokine/immunology , Th2 Cells/immunology , Animals , Antibodies, Monoclonal/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Gene Expression Regulation/drug effects , Interleukin-4/pharmacology , Mice , Mice, Inbred BALB C , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, CCR4 , Receptors, CXCR3 , Receptors, Chemokine/drug effects , Receptors, Chemokine/genetics , STAT6 Transcription Factor/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...