Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Cell ; 139(6): 1069-83, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19962179

ABSTRACT

Chromosomal translocations are a hallmark of leukemia/lymphoma and also appear in solid tumors, but the underlying mechanism remains elusive. By establishing a cellular model that mimics the relative frequency of authentic translocation events without proliferation selection, we report mechanisms of nuclear receptor-dependent tumor translocations. Intronic binding of liganded androgen receptor (AR) first juxtaposes translocation loci by triggering intra- and interchromosomal interactions. AR then promotes site-specific DNA double-stranded breaks (DSBs) at translocation loci by recruiting two types of enzymatic activities induced by genotoxic stress and liganded AR, including activation-induced cytidine deaminase and the LINE-1 repeat-encoded ORF2 endonuclease. These enzymes synergistically generate site-selective DSBs at juxtaposed translocation loci that are ligated by nonhomologous end joining pathway for specific translocations. Our data suggest that the confluence of two parallel pathways initiated by liganded nuclear receptor and genotoxic stress underlies nonrandom tumor translocations, which may function in many types of tumors and pathological processes.


Subject(s)
Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism , Transcription, Genetic , Translocation, Genetic , Cell Line, Tumor , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Introns , Long Interspersed Nucleotide Elements , Male , Open Reading Frames , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Regulator ERG
2.
Cell ; 132(6): 996-1010, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18358812

ABSTRACT

While the transcriptional machinery has been extensively dissected at the molecular level, little is known about regulation of chromosomal organization in the three-dimensional space of the nucleus to achieve integrated transcriptional responses to diverse signaling events. Here, we report that ligand induces rapid interchromosomal interactions among subsets of estrogen receptor alpha-bound transcription units, with a dramatic reorganization of nuclear territories requiring nuclear actin/myosin-I transport machinery, dynein light chain 1 (DLC1), and a specific subset of transcriptional coactivators and chromatin remodeling complexes. We establish a requirement for the histone lysine demethylase, LSD1, in directing specific interchromosomal interaction loci to distinct interchromatin granules, long thought to be "storage" sites for splicing machinery, and demonstrate that these three-dimensional motor-dependent interactions are required to achieve enhanced transcription of specific estrogen-receptor target genes. These findings reveal roles for the modulation of nuclear architecture in orchestrating regulated gene-expression programs in the mammalian nucleus.


Subject(s)
Chromatin/metabolism , Estrogen Receptor alpha/metabolism , Gene Regulatory Networks , Molecular Motor Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , Actins/metabolism , Cell Line, Tumor , Cell Nucleus , Cells, Cultured , Histone Demethylases , Humans , Intranuclear Inclusion Bodies/metabolism , Transcription, Genetic
5.
Mol Cell ; 29(6): 755-66, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18374649

ABSTRACT

A key strategy to achieve regulated gene expression in higher eukaryotes is to prevent illegitimate signal-independent activation by imposing robust control on the dismissal of corepressors. Here, we report that many signaling pathways, including Notch, NF-kappaB, and nuclear receptor ligands, are subjected to a dual-repression "checkpoint" based on distinct corepressor complexes. Gene activation requires the release of both CtBP1/2- and NCoR/SMRT-dependent repression, through the coordinate action of two highly related exchange factors, the transducer beta-like proteins TBL1 and TBLR1, that license ubiquitylation and degradation of CtBP1/2 and NCoR/SMRT, respectively. Intriguingly, their function and differential specificity reside in only five specific Ser/Thr phosphorylation site differences, regulated by direct phosphorylation at the level of the promoter, as exemplified by the role of PKCdelta in TBLR1-dependent dismissal of NCoR. Thus, our data reveal a strategy of dual-factor repression checkpoints, in which dedicated exchange factors serve as sensors for signal-specific dismissal of distinct corepressors, with specificity imposed by upstream signaling pathways.


Subject(s)
Alcohol Oxidoreductases/genetics , DNA-Binding Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Transducin/metabolism , Animals , Breast Neoplasms , Cell Line , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Nuclear Receptor Co-Repressor 2 , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/genetics , Transcriptional Activation , Ubiquitin/metabolism
6.
Nature ; 434(7035): 921-6, 2005 Apr 14.
Article in English | MEDLINE | ID: mdl-15829968

ABSTRACT

Defining the molecular strategies that integrate diverse signalling pathways in the expression of specific gene programmes that are critical in homeostasis and disease remains a central issue in biology. This is particularly pertinent in cancer biology because downregulation of tumour metastasis suppressor genes is a common occurrence, and the underlying molecular mechanisms are not well established. Here we report that the downregulation of a metastasis suppressor gene, KAI1, in prostate cancer cells involves the inhibitory actions of beta-catenin, along with a reptin chromatin remodelling complex. This inhibitory function of beta-catenin-reptin requires both increased beta-catenin expression and recruitment of histone deacetylase activity. The coordinated actions of beta-catenin-reptin components that mediate the repressive state serve to antagonize a Tip60 coactivator complex that is required for activation; the balance of these opposing complexes controls the expression of KAI1 and metastatic potential. The molecular mechanisms underlying the antagonistic regulation of beta-catenin-reptin and the Tip60 coactivator complexes for the metastasis suppressor gene, KAI1, are likely to be prototypic of a selective downregulation strategy for many genes, including a subset of NF-kappaB target genes.


Subject(s)
Acetyltransferases/metabolism , Antigens, CD/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Membrane Glycoproteins/genetics , Neoplasm Metastasis/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/metabolism , Transcription, Genetic/genetics , Acetyltransferases/genetics , Animals , Cell Line, Tumor , Chromatin Assembly and Disassembly , Collagen , Down-Regulation/genetics , Drug Combinations , Histone Acetyltransferases , Humans , Kangai-1 Protein , Laminin , Lysine Acetyltransferase 5 , Male , Mice , NF-kappa B/metabolism , Neoplasm Transplantation , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/metabolism , Proteoglycans , RNA, Messenger/genetics , RNA, Messenger/metabolism , beta Catenin
7.
Nature ; 437(7059): 759-63, 2005 Sep 29.
Article in English | MEDLINE | ID: mdl-16127449

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) has essential roles in adipogenesis and glucose homeostasis, and is a molecular target of insulin-sensitizing drugs. Although the ability of PPAR-gamma agonists to antagonize inflammatory responses by transrepression of nuclear factor kappa B (NF-kappaB) target genes is linked to antidiabetic and antiatherogenic actions, the mechanisms remain poorly understood. Here we report the identification of a molecular pathway by which PPAR-gamma represses the transcriptional activation of inflammatory response genes in mouse macrophages. The initial step of this pathway involves ligand-dependent SUMOylation of the PPAR-gamma ligand-binding domain, which targets PPAR-gamma to nuclear receptor corepressor (NCoR)-histone deacetylase-3 (HDAC3) complexes on inflammatory gene promoters. This in turn prevents recruitment of the ubiquitylation/19S proteosome machinery that normally mediates the signal-dependent removal of corepressor complexes required for gene activation. As a result, NCoR complexes are not cleared from the promoter and target genes are maintained in a repressed state. This mechanism provides an explanation for how an agonist-bound nuclear receptor can be converted from an activator of transcription to a promoter-specific repressor of NF-kappaB target genes that regulate immunity and homeostasis.


Subject(s)
Down-Regulation , Inflammation/genetics , PPAR gamma/metabolism , Repressor Proteins/metabolism , SUMO-1 Protein/metabolism , Animals , Cells, Cultured , Down-Regulation/drug effects , Histone Deacetylases/metabolism , Ligands , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Protein Binding/drug effects , Protein Inhibitors of Activated STAT , Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 105(7): 2481-6, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18272476

ABSTRACT

Enhancers have been functionally described for >35 years, but the molecular principles underlying the integration of regulatory inputs to alternate gene enhancers used during mammalian organogenesis remain incompletely understood. Using a combination of in vivo enhancer mapping and proteomics approaches, we have established that two distant and distinct early enhancers, each requiring different transcription complexes, are required for full activation of the gene encoding the pituitary lineage determining factor, Pit1. A transcription factor belonging to the "giant, multiple-homeodomain and zinc finger family," Atbf1, serves as a novel pituitary regulator for one of the two required enhancers as shown by genetic and in vitro analysis.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Transcription Factor Pit-1/metabolism , Animals , Base Sequence , Cell Lineage , Embryonic Stem Cells/metabolism , Epistasis, Genetic , Genome/genetics , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation/genetics , Pituitary Gland/metabolism , Protein Binding , Proteomics , Time Factors , Transcription Factor Pit-1/genetics
9.
Proc Natl Acad Sci U S A ; 105(49): 19199-204, 2008 12 09.
Article in English | MEDLINE | ID: mdl-19052240

ABSTRACT

Although the role of liganded nuclear receptors in mediating coactivator/corepressor exchange is well-established, little is known about the potential regulation of chromosomal organization in the 3-dimensional space of the nucleus in achieving integrated transcriptional responses to diverse signaling events. Here, we report that ligand induces rapid interchromosomal interactions among specific subsets of estrogen receptor alpha-bound transcription units, with a dramatic reorganization of nuclear territories, which depends on the actions of nuclear actin/myosin-I machinery and dynein light chain 1. The histone lysine demethylase, LSD1, is required for these ligand-induced interactive loci to associate with distinct interchromatin granules, long thought to serve as "storage" sites for the splicing machinery, some critical transcription elongation factors, and various chromatin remodeling complexes. We demonstrate that this 2-step nuclear rearrangement is essential for achieving enhanced, coordinated transcription of nuclear receptor target genes.


Subject(s)
Epithelial Cells/physiology , Gene Regulatory Networks/physiology , Oxidoreductases, N-Demethylating/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription, Genetic/physiology , Breast Neoplasms , Cell Line, Tumor , Cell Nucleus/physiology , Chromatin/physiology , Epithelial Cells/cytology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Gene Expression Regulation/physiology , Histone Demethylases , Humans , In Situ Hybridization, Fluorescence , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidoreductases, N-Demethylating/chemistry , Oxidoreductases, N-Demethylating/metabolism , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/metabolism , Trefoil Factor-1 , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
10.
Nature ; 426(6964): 247-54, 2003 Nov 20.
Article in English | MEDLINE | ID: mdl-14628042

ABSTRACT

The precise mechanistic relationship between gene activation and repression events is a central question in mammalian organogenesis, as exemplified by the evolutionarily conserved sine oculis (Six), eyes absent (Eya) and dachshund (Dach) network of genetically interacting proteins. Here, we report that Six1 is required for the development of murine kidney, muscle and inner ear, and that it exhibits synergistic genetic interactions with Eya factors. We demonstrate that the Eya family has a protein phosphatase function, and that its enzymatic activity is required for regulating genes encoding growth control and signalling molecules, modulating precursor cell proliferation. The phosphatase function of Eya switches the function of Six1-Dach from repression to activation, causing transcriptional activation through recruitment of co-activators. The gene-specific recruitment of a co-activator with intrinsic phosphatase activity provides a molecular mechanism for activation of specific gene targets, including those regulating precursor cell proliferation and survival in mammalian organogenesis.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Organogenesis , Protein Tyrosine Phosphatases/metabolism , Trans-Activators/metabolism , Animals , Cell Division , Cell Survival , DNA-Binding Proteins/genetics , Ear/embryology , Gene Deletion , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Kidney/embryology , Kidney/metabolism , Mice , Mice, Knockout , Muscles/embryology , Muscles/metabolism , Pituitary Gland/embryology , Pituitary Gland/metabolism , Protein Binding , Protein Tyrosine Phosphatases/genetics , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/genetics , Transcriptional Activation
11.
Am J Physiol Renal Physiol ; 297(5): F1330-41, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19726549

ABSTRACT

Kidney organogenesis depends on reciprocal interactions between the ureteric bud (UB) and the metanephric mesenchyme (MM) to form the UB-derived collecting system and MM-derived nephron. With the advent of in vitro systems, it is clear that UB branching can occur independently of MM contact; however, little has been done to detail the role of MM cellular contact in this process. Here, a model system in which the cultured isolated UB is recombined with uninduced MM is used to isolate the effects of the MM progenitor tissue on the development and maturation of the collecting system. By morphometrics, we demonstrate that cellular contact with the MM is required for vectorial elongation of stalks and tapering of luminal caliber of UB-derived tubules. Expression analysis of developmentally significant genes indicates the cocultured tissue is most similar to an embryonic day 19 (E19) kidney. The likely major contributor to this is the functional maturation of the collecting duct and proximal nephron segments in the UB-induced MM, as measured by quantitative PCR, of the collecting duct-specific arginine vasopressin receptor and the nephron tubule segment-specific organic anion transporter OAT1, Na-P(i) type 2 cotransporter, and Tamm-Horsfall protein gene expressions. However, expression of aquaporin-2 is upregulated similarly in isolated UB and cocultured tissue, suggesting that some aspects of functional maturation can occur independently of MM cellular contact. In addition to its sculpting effects, the MM normalized a "branchless" UB morphology induced by FGF7 or heregulin in isolated UB culture. The morphological changes induced by the MM were accompanied by a reassignment of GFRalpha1 (a receptor for GDNF) to tips. Such "quality control" by the MM of UB morphology may provide resiliency to the branching program. This may help to explain a number of knockout phenotypes in which branching and/or cystic defects are less impressive than expected. A second hit in the MM may thus be necessary to make these defects fully apparent.


Subject(s)
Kidney/embryology , Mesoderm/physiology , Uterus/abnormalities , Uterus/embryology , Animals , Coculture Techniques , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Female , Fluorescent Dyes , Immunohistochemistry , Kidney/anatomy & histology , Microarray Analysis , Microinjections , Phenotype , Pregnancy , Rats , Reverse Transcriptase Polymerase Chain Reaction , Rhodamines , Uterus/anatomy & histology
12.
Mol Cell Biol ; 26(4): 1386-97, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16449650

ABSTRACT

The transcriptional corepressor mSin3 is associated with histone deacetylases (HDACs) and is utilized by many DNA-binding transcriptional repressors. We have cloned and characterized a novel mSin3A-binding protein, SAP25. SAP25 binds to the PAH1 domain of mSin3A, associates with the mSin3A-HDAC complex in vivo, and represses transcription when tethered to DNA. SAP25 is required for mSin3A-mediated, but not N-CoR-mediated, repression. SAP25 is a nucleocytoplasmic shuttling protein, actively exported from the nucleus by a CRM1-dependent mechanism. A fraction of SAP25 is located in promyelocytic leukemia protein (PML) nuclear bodies, and PML induces a striking nuclear accumulation of SAP25. An isotope-coded affinity tag quantitative proteomic analysis of the SAP25 complex revealed that SAP25 is associated with several components of the mSin3 complex, nuclear export machinery, and regulators of transcription and cell cycle. These results suggest that SAP25 is a novel core component of the mSin3 corepressor complex whose subcellular location is regulated by PML.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Amino Acid Sequence , Animals , BALB 3T3 Cells , Carrier Proteins/genetics , Cell Line , Cloning, Molecular , HeLa Cells , Histone Deacetylases/genetics , Humans , In Vitro Techniques , Mice , Molecular Sequence Data , Multiprotein Complexes , Rats , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
13.
Neuron ; 40(6): 1119-31, 2003 Dec 18.
Article in English | MEDLINE | ID: mdl-14687547

ABSTRACT

The cerebellum provides an excellent system for understanding how afferent and target neurons coordinate sequential intercellular signals and cell-autonomous genetic programs in development. Mutations in the orphan nuclear receptor RORalpha block Purkinje cell differentiation with a secondary loss of afferent granule cells. We show that early transcriptional targets of RORalpha include both mitogenic signals for afferent progenitors and signal transduction genes required to process their subsequent synaptic input. RORalpha acts through recruitment of gene-specific sets of transcriptional cofactors, including beta-catenin, p300, and Tip60, but appears independent of CBP. One target promoter is Sonic hedgehog, and recombinant Sonic hedgehog restores granule precursor proliferation in RORalpha-deficient cerebellum. Our results suggest a link between RORalpha and beta-catenin pathways, confirm that a nuclear receptor employs distinct coactivator complexes at different target genes, and provide a logic for early RORalpha expression in coordinating expression of genes required for reciprocal signals in cerebellar development.


Subject(s)
Calcium Signaling/physiology , Cerebellum/growth & development , Cerebellum/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Trans-Activators/biosynthesis , Animals , Hedgehog Proteins , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nuclear Receptor Subfamily 1, Group F, Member 1 , Purkinje Cells/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Trans-Activators/genetics
14.
Immunol Res ; 38(1-3): 51-4, 2007.
Article in English | MEDLINE | ID: mdl-17917009

ABSTRACT

Dr. Robert A. Good and the March of Dimes Birth Defects Foundation maintained a close association for a quarter century in the fight against immunodeficiency diseases. The March of Dimes, whose mission is to prevent birth defects, premature birth, and infant mortality, awarded an initial grant to Dr. Good in 1960 to conduct basic clinical and experimental studies on arthritis and collagen diseases. By 1966, this support broadened to include Dr. Good's research on agammaglobulinemia, ataxia telangiectasia, Chediak-Higashi disease, and Wiskott-Aldrich syndrome. Dr. Good led three historic March of Dimes conferences on immunodeficiency and, in 1968, conducted the first bone marrow transplant to correct an immunological birth defect, memorialized by the March of Dimes in its educational film, Decision (1970). March of Dimes grants to Dr. Good for his research in cellular engineering to genetically correct the defined birth defects approached $1 million for the period 1960-1985.


Subject(s)
Foundations/history , Immunologic Deficiency Syndromes/history , History, 20th Century , History, 21st Century , Humans , Immunologic Deficiency Syndromes/economics , United States
15.
Mol Cell Biol ; 22(17): 6272-85, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12167719

ABSTRACT

beta-Arrestin-1 mediates agonist-dependent desensitization and internalization of G protein-coupled receptors (GPCRs) and is also essential for GPCR mitogenic signaling. In addition, insulin-like growth factor I receptor (IGF-IR) endocytosis is facilitated by beta-arrestin-1, and internalization is necessary for IGF-I-stimulated mitogen-activated protein (MAP) kinase activation. Here, we report that treatment of cells for 12 h with insulin (100 ng/ml) induces an approximately 50% decrease in cellular beta-arrestin-1 content due to ubiquitination of beta-arrestin-1 and proteosome-mediated degradation. This insulin-induced decrease in beta-arrestin-1 content was blocked by inhibition of phosphatidylinositol-3 kinase (PI-3 kinase) and MEK with wortmannin and PD98059, respectively. We also found a marked decrease in the association of beta-arrestin-1 with the IGF-IR and a 55% inhibition of IGF-I-stimulated MAP kinase phosphorylation. In insulin-treated, beta-arrestin-1-downregulated cells, there was complete inhibition of lysophosphatidic acid (LPA) or isoproterenol (ISO)-stimulated MAP kinase phosphorylation. This was associated with a decrease in beta-arrestin-1 association with the beta2-AR as well as a decrease in beta-arrestin-1-Src and Src-beta2-AR association. Ectopic expression of wild-type beta-arrestin-1 in insulin-treated cells in which endogenous beta-arrestin-1 had been downregulated rescued IGF-I- and LPA-stimulated MAP kinase phosphorylation. In conclusion, we found the following. (i) Chronic insulin treatment leads to enhanced beta-arrestin-1 degradation. (ii) This downregulation of endogenous beta-arrestin-1 is associated with decreased IGF-I-, LPA-, and ISO-mediated MAP kinase signaling, which can be rescued by ectopic expression of wild-type beta-arrestin-1. (iii) Finally, these results describe a novel mechanism for heterologous desensitization, whereby insulin treatment can impair GPCR signaling, and highlight the importance of beta-arrestin-1 as a target molecule for this desensitization mechanism.


Subject(s)
Arrestins/metabolism , GTP-Binding Proteins/physiology , Insulin-Like Growth Factor I/pharmacology , Insulin/pharmacology , Proteasome Endopeptidase Complex , Receptors, Adrenergic, beta-2/drug effects , Signal Transduction/drug effects , 3T3 Cells/drug effects , Adipocytes/drug effects , Adrenergic beta-Agonists/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Humans , Isoproterenol/pharmacology , Lysophospholipids/pharmacology , MAP Kinase Kinase 1 , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Peptide Hydrolases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Receptor, IGF Type 1/metabolism , Receptor, Insulin/drug effects , Receptor, Insulin/physiology , Receptors, Adrenergic, beta-2/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/physiology , Signal Transduction/physiology , Ubiquitin/metabolism , beta-Arrestin 1 , beta-Arrestins
16.
Mol Cell Biol ; 22(12): 4319-33, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12024042

ABSTRACT

The aryl hydrocarbon receptor complex heterodimeric transcription factor, comprising the basic helix-loop-helix-Per-ARNT-Sim (bHLH-PAS) domain aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) proteins, mediates the toxic effects of TCDD (2,3,7,8 tetrachlorodibenzo-p-dioxin). The molecular events underlying TCDD-inducible gene activation, beyond the activation of the AHRC, are poorly understood. The SRC-1/NCoA-1, NCoA-2/GRIP-1/TIF-2, and p/CIP/AIB/ACTR proteins have been shown to act as mediators of transcriptional activation. In this report, we demonstrate that SRC-1, NCoA-2, and p/CIP are capable of independently enhancing TCDD-dependent induction of a luciferase reporter gene by the AHR/ARNT dimer. Furthermore, injection of anti-SRC-1 or anti-p/CIP immunoglobulin G into mammalian cells abolishes the transcriptional activity of a TCDD-dependent reporter gene. We demonstrate by coimmunoprecipitation and by a reporter gene assay that SRC-1 and NCoA-2 but not p/CIP are capable of interacting with ARNT in vivo after transient transfection into mammalian cells, while AHR is capable of interacting with all three coactivators. We confirm the interactions of ARNT and AHR with SRC-1 with immunocytochemical techniques. Furthermore, SRC-1, NCoA-2, and p/CIP all associate with the CYP1A1 enhancer region in a TCDD-dependent fashion, as demonstrated by chromatin immunoprecipitation assays. We demonstrate by yeast two-hybrid, glutathione S-transferase pulldown, and mammalian reporter gene assays that ARNT requires its helix 2 domain but not its transactivation domain to interact with SRC-1. This indicates a novel mechanism of action for SRC-1. SRC-1 does not require its bHLH-PAS domain to interact with ARNT or AHR, but utilizes distinct domains proximal to its p300/CBP interaction domain. Taken together, these data support a role for the SRC family of transcriptional coactivators in TCDD-dependent gene regulation.


Subject(s)
DNA-Binding Proteins , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator , Cells, Cultured , Cytochrome P-450 CYP1A1/genetics , Enhancer Elements, Genetic , Gene Expression Regulation/drug effects , Helix-Loop-Helix Motifs , Histone Acetyltransferases , Humans , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2 , Nuclear Receptor Coactivator 3 , Polychlorinated Dibenzodioxins/toxicity , Precipitin Tests , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/genetics , Teratogens/toxicity , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/drug effects , Transcription Factors/genetics , Transcriptional Activation
17.
Endocrinology ; 143(2): 655-64, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11796522

ABSTRACT

Previously, we had shown that inhibition of PLC activity impaired the ability of insulin to activate ERK in 3T3-L1 adipocytes. In this study, we confirmed that the insulin receptor and PLC-gamma1 are physically associated in hIRcB fibroblasts, insulin stimulates PLC-gamma1 enzyme activity, and inhibition of PLC activity impairs activation of ERK. We subsequently investigated whether PLC-gamma1 is required for insulin-stimulated mitogenesis. First, inhibition of PLC activity using U73122 impairs the ability of insulin to stimulate DNA synthesis. Second, disruption of the interaction of the insulin receptor with PLC-gamma1 by microinjection of SH2 domains derived from PLC-gamma1 or Grb2 but not Shc similarly blocks insulin-induced DNA synthesis. Third, microinjection of neutralizing antibodies to PLC-gamma1 blocks DNA synthesis, but nonneutralizing antibodies do not. The blockade in all three cases is rescued by synthetic diacylglycerols but not by inositol-1,4,5-trisphosphate, indicating a requirement for PLC enzyme activity. These experimental data point to a requirement for PLC-gamma1 in insulin-stimulated mitogenesis in hIRcB cells.


Subject(s)
DNA/biosynthesis , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Isoenzymes/metabolism , Type C Phospholipases/metabolism , src Homology Domains , Animals , Cell Line , Fibroblasts , Gene Expression Regulation, Enzymologic/genetics , Glutathione Transferase/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phospholipase C gamma , Phosphorylation , Rats , Receptor, Insulin/drug effects , Recombinant Fusion Proteins , Signal Transduction/drug effects , Stimulation, Chemical , Type C Phospholipases/antagonists & inhibitors
18.
Nat Struct Mol Biol ; 19(11): 1168-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23064648

ABSTRACT

Although liganded nuclear receptors have been established to regulate RNA polymerase II (Pol II)-dependent transcription units, their role in regulating Pol III-transcribed DNA repeats remains largely unknown. Here we report that ~2-3% of the ~100,000-200,000 total human DR2 Alu repeats located in proximity to activated Pol II transcription units are activated by the retinoic acid receptor (RAR) in human embryonic stem cells to generate Pol III-dependent RNAs. These transcripts are processed, initially in a DICER-dependent fashion, into small RNAs (~28-65 nt) referred to as repeat-induced RNAs that cause the degradation of a subset of crucial stem-cell mRNAs, including Nanog mRNA, which modulate exit from the proliferative stem-cell state. This regulation requires AGO3-dependent accumulation of processed DR2 Alu transcripts and the subsequent recruitment of AGO3-associated decapping complexes to the target mRNA. In this way, the RAR-dependent and Pol III-dependent DR2 Alu transcriptional events in stem cells functionally complement the Pol II-dependent neuronal transcriptional program.


Subject(s)
Argonaute Proteins/metabolism , DEAD-box RNA Helicases/metabolism , Embryonic Stem Cells/physiology , RNA, Small Interfering/metabolism , Receptors, Retinoic Acid/metabolism , Ribonuclease III/metabolism , Transcription, Genetic/physiology , Alu Elements/genetics , Alu Elements/physiology , Base Sequence , Blotting, Northern , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , DNA Polymerase III/physiology , Embryonic Stem Cells/metabolism , Humans , In Situ Hybridization, Fluorescence , Mass Spectrometry , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
19.
Mol Biol Cell ; 19(6): 2579-87, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18385516

ABSTRACT

GRASP55 is a Golgi-associated protein, but its function at the Golgi remains unclear. Addition of full-length GRASP55, GRASP55-specific peptides, or an anti-GRASP55 antibody inhibited Golgi fragmentation by mitotic extracts in vitro, and entry of cells into mitosis. Phospho-peptide mapping of full-length GRASP55 revealed that threonine 225 and 249 were mitotically phosphorylated. Wild-type peptides containing T225 and T249 inhibited Golgi fragmentation and entry of cells into mitosis. Mutant peptides containing T225E and T249E, in contrast, did not affect Golgi fragmentation and entry into mitosis. These findings reveal a role of GRASP55 in events leading to Golgi fragmentation and the subsequent entry of cell into mitosis. Surprisingly, however, under our experimental conditions, >85% knockdown of GRASP55 did not affect the overall organization of Golgi organization in terms of cisternal stacking and lateral connections between stacks. Based on our findings we suggest that phosphorylation of GRASP55 at T225/T249 releases a bound component, which is phosphorylated and necessary for Golgi fragmentation. Thus, GRASP55 has no role in the organization of Golgi membranes per se, but it controls their fragmentation by regulating the release of a partner, which requires a G2-specific phosphorylation at T225/T249.


Subject(s)
Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Mitosis , Amino Acid Sequence , Animals , Antibody Specificity , Cell Extracts , Cell Line , Cloning, Molecular , Golgi Apparatus/ultrastructure , Golgi Matrix Proteins , Humans , Membrane Proteins/chemistry , Molecular Sequence Data , Peptide Mapping , Phosphoproteins/metabolism , Phosphorylation , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Rats
20.
CSH Protoc ; 2007: pdb.prot4754, 2007 May 01.
Article in English | MEDLINE | ID: mdl-21357085

ABSTRACT

INTRODUCTIONMicroinjection allows the introduction of molecules into a defined population of cells at a defined concentration, and the timing of the experiment can be controlled stringently, minimizing problems associated with overexpression. Delivery by microinjection can be used for any type of cell that is adherent in culture, including primary cells. Perhaps its most powerful aspect is the ability to introduce several types of reagents into cells simultaneously, including DNA constructs, a labeled dextran to mark injected cells, antibodies, short interfering RNAs (siRNAs), and peptides. Because siRNA can be generated easily and rapidly for any target gene, it is relatively simple to assess many effects of the knockout of any gene in any type of adherent cell in a matter of days. However, microinjection is not trivial to learn, and requires attention to detail. Also, the limited number of cells involved often does not permit the subsequent analysis of effects on some biochemical parameters. While the major limitation of the approach is the small amount of material obtained, as the sensitivity of analysis techniques increases, so does the usefulness of this method. We emphasize here recent advances in the use of microinjection in combination with siRNA technology.

SELECTION OF CITATIONS
SEARCH DETAIL