Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
2.
Genes Dev ; 15(23): 3193-207, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11731482

ABSTRACT

Understanding the functional significance of the coordinate expression of specific corepressors and DNA-binding transcription factors remains a critical question in mammalian development. During the development of the pituitary gland, two highly related paired-like homeodomain factors, a repressor, Hesx1/Rpx and an activator, Prop-1, are expressed in sequential, overlapping temporal patterns. Here we show that while the repressive actions of Hesx1/Rpx may be required for initial pituitary organ commitment, progression beyond the appearance of the first pituitary (POMC) lineage requires both loss of Hesx1 expression and the actions of Prop-1. Although Hesx1 recruits both the Groucho-related corepressor TLE1 and the N-CoR/Sin3/HDAC complex on distinct domains, the repressor functions of Hesx1 in vivo prove to require the specific recruitment of TLE1, which exhibits a spatial and temporal pattern of coexpression during pituitary organogenesis. Furthermore, Hesx1-mediated repression coordinates a negative feedback loop with FGF8/FGF10 signaling in the ventral diencephalon, required to prevent induction of multiple pituitary glands from oral ectoderm. Our data suggest that the opposing actions of two structurally-related DNA-binding paired-like homeodomain transcription factors, binding to similar cognate elements, coordinate pituitary organogenesis by reciprocally repressing and activating target genes in a temporally specific fashion, on the basis of the actions of a critical, coexpressed TLE corepressor.


Subject(s)
Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Pituitary Gland/embryology , Repressor Proteins/metabolism , Trans-Activators/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Cell Lineage , Co-Repressor Proteins , Evolution, Molecular , Feedback, Physiological , Fibroblast Growth Factors/metabolism , HeLa Cells , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Macromolecular Substances , Mice , Mice, Transgenic , Models, Biological , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pituitary Gland/cytology , Pituitary Gland/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Repressor Proteins/chemistry , Repressor Proteins/genetics , Trans-Activators/chemistry , Trans-Activators/genetics , Transcription Factor HES-1
3.
Mol Cell Biol ; 21(21): 7207-17, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11585904

ABSTRACT

The Raf kinase inhibitor protein (RKIP) acts as a negative regulator of the mitogen-activated protein (MAP) kinase (MAPK) cascade initiated by Raf-1. RKIP inhibits the phosphorylation of MAP/extracellular signal-regulated kinase 1 (MEK1) by Raf-1 by disrupting the interaction between these two kinases. We show here that RKIP also antagonizes the signal transduction pathways that mediate the activation of the transcription factor nuclear factor kappa B (NF-kappaB) in response to stimulation with tumor necrosis factor alpha (TNF-alpha) or interleukin 1 beta. Modulation of RKIP expression levels affected NF-kappaB signaling independent of the MAPK pathway. Genetic epistasis analysis involving the ectopic expression of kinases acting in the NF-kappaB pathway indicated that RKIP acts upstream of the kinase complex that mediates the phosphorylation and inactivation of the inhibitor of NF-kappaB (IkappaB). In vitro kinase assays showed that RKIP antagonizes the activation of the IkappaB kinase (IKK) activity elicited by TNF-alpha. RKIP physically interacted with four kinases of the NF-kappaB activation pathway, NF-kappaB-inducing kinase, transforming growth factor beta-activated kinase 1, IKKalpha, and IKKbeta. This mode of action bears striking similarities to the interactions of RKIP with Raf-1 and MEK1 in the MAPK pathway. Emerging data from diverse organisms suggest that RKIP and RKIP-related proteins represent a new and evolutionarily highly conserved family of protein kinase regulators. Since the MAPK and NF-kappaB pathways have physiologically distinct roles, the function of RKIP may be, in part, to coordinate the regulation of these pathways.


Subject(s)
Androgen-Binding Protein , Carrier Proteins/metabolism , Carrier Proteins/physiology , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Animals , COS Cells , Cell Line , Enzyme Activation , Evolution, Molecular , Genes, Reporter , Humans , Interleukin-1/metabolism , Kinetics , Phosphatidylethanolamine Binding Protein , Phospholipid Transfer Proteins , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Prostatein , Protein Binding , Protein Structure, Tertiary , Rats , Secretoglobins , Signal Transduction , Transfection , Tumor Necrosis Factor-alpha/metabolism , Uteroglobin , NF-kappaB-Inducing Kinase
4.
Neuron ; 31(3): 353-65, 2001 Aug 16.
Article in English | MEDLINE | ID: mdl-11516394

ABSTRACT

The transcriptional repressor, REST, helps restrict neuronal traits to neurons by blocking their expression in nonneuronal cells. To examine the repercussions of REST expression in neurons, we generated a neuronal cell line that expresses REST conditionally. REST expression inhibited differentiation by nerve growth factor, suppressing both sodium current and neurite growth. A novel corepressor complex, CoREST/HDAC2, was shown to be required for REST repression. In the presence of REST, the CoREST/HDAC2 complex occupied the native Nav1.2 sodium channel gene in chromatin. In neuronal cells that lack REST and express sodium channels, the corepressor complex was not present on the gene. Collectively, these studies define a novel HDAC complex that is recruited by the C-terminal repressor domain of REST to actively repress genes essential to the neuronal phenotype.


Subject(s)
Cerebral Cortex/physiology , Neurons/physiology , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , COS Cells , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Chlorocebus aethiops , Chromatin/physiology , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Histone Deacetylase 2 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice , Mice, Inbred C57BL , NAV1.2 Voltage-Gated Sodium Channel , Nerve Growth Factor/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neurons/cytology , Neurons/drug effects , PC12 Cells , Rats , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Sodium Channels/genetics , Sodium Channels/physiology , Transcription Factors/genetics , Transfection , Zinc Fingers
5.
Blood ; 96(10): 3537-43, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071652

ABSTRACT

Adenine deoxynucleosides induce apoptosis in quiescent lymphocytes and are thus useful drugs for the treatment of indolent lymphoproliferative diseases. To explain why deoxyadenosine and its analogs are toxic to a cell that is not undergoing replicative DNA synthesis, several mechanisms have been proposed, including the direct binding of dATP to the pro-apoptotic factor Apaf-1 and the activation of the caspase-9 and -3 pathways. In this study it is shown, by means of several assays on whole cells and isolated mitochondria, that 2-chloro-2'-deoxyadenosine (2CdA) and 2-choloro-2'-ara-fluorodeoxyadenosine (CaFdA) disrupt the integrity of mitochondria from primary chronic lymphocytic leukemia (B-CLL) cells. The nucleoside-induced damage leads to the release of the pro-apoptotic mitochondrial proteins cytochrome c and apoptosis-inducing factor. The other adenine deoxynucleosides tested displayed comparable DNA-damaging potency but did not affect mitochondrial function. Interference with mitochondrial integrity, thus, may be a factor in the potent cytotoxic effects of 2CdA and CaFdA toward nondividing lymphocytes.


Subject(s)
Apoptosis/drug effects , DNA Damage/physiology , Deoxyadenosines/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mitochondria/drug effects , Vidarabine/analogs & derivatives , Adenine Nucleotides , Adenosine Triphosphate/administration & dosage , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Antineoplastic Agents/pharmacology , Arabinonucleosides/pharmacology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/ultrastructure , Cell Survival/drug effects , Cladribine/pharmacology , Clofarabine , Comet Assay , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Deoxyadenosines/physiology , Humans , Immunohistochemistry , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Membrane Potentials/drug effects , Microinjections , Mitochondria/physiology , Mitochondria/ultrastructure , Time Factors , Tumor Cells, Cultured , Vidarabine/pharmacology
6.
Science ; 290(5494): 1127-31, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11073444

ABSTRACT

Reciprocal gene activation and restriction during cell type differentiation from a common lineage is a hallmark of mammalian organogenesis. A key question, then, is whether a critical transcriptional activator of cell type-specific gene targets can also restrict expression of the same genes in other cell types. Here, we show that whereas the pituitary-specific POU domain factor Pit-1 activates growth hormone gene expression in one cell type, the somatotrope, it restricts its expression from a second cell type, the lactotrope. This distinction depends on a two-base pair spacing in accommodation of the bipartite POU domains on a conserved growth hormone promoter site. The allosteric effect on Pit-1, in combination with other DNA binding factors, results in the recruitment of a corepressor complex, including nuclear receptor corepressor N-CoR, which, unexpectedly, is required for active long-term repression of the growth hormone gene in lactotropes.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Gene Expression Regulation , Growth Hormone/genetics , Pituitary Gland/metabolism , Prolactin/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Allosteric Regulation , Animals , Base Sequence , Binding Sites , Cell Line , Conserved Sequence , Crystallization , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Female , Genes, Reporter , Male , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Pituitary Gland/cytology , Promoter Regions, Genetic , Protein Conformation , Protein Structure, Tertiary , Rats , Repressor Proteins/chemistry , Repressor Proteins/genetics , Transcription Factor Pit-1 , Transcription Factors/chemistry , Transcription Factors/genetics , Transcriptional Activation
7.
Proc Natl Acad Sci U S A ; 97(25): 13549-54, 2000 Dec 05.
Article in English | MEDLINE | ID: mdl-11087842

ABSTRACT

A family of p160 coactivators was initially identified based on ligand-dependent interactions with nuclear receptors and thought to function, in part, by recruiting CREB-binding protein/p300 to several classes of transcription factors. One of the p160 factors, p/CIP/AIB1, often amplified and overexpressed in breast cancer, also exhibits particularly strong interaction with CREB-binding protein/p300. In this manuscript, we report that p/CIP, which exhibits regulated transfer from cytoplasm to nucleus, is required for normal somatic growth from embryonic day 13.5 through maturity. Our data suggest that a short stature phenotype of p/CIP gene-deleted mice reflect both altered regulation of insulin-like growth factor-1 (IGF-1) gene expression in specific tissues and a cell-autonomous defect of response to IGF-1, including ineffective transcriptional activities by several classes of regulated transcription factors under specific conditions. The actions of p/CIP are therefore required for full expression of a subset of genes critical for regulating physiological patterns of somatic growth in mammals.


Subject(s)
Cell Division/physiology , Trans-Activators/physiology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Female , Gene Deletion , Mice , Mice, Inbred C57BL , Trans-Activators/genetics
8.
Cell ; 102(6): 753-63, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-11030619

ABSTRACT

Transcriptional repression plays crucial roles in diverse aspects of metazoan development, implying critical regulatory roles for corepressors such as N-CoR and SMRT. Altered patterns of transcription in tissues and cells derived from N-CoR gene-deleted mice and the resulting block at specific points in CNS, erythrocyte, and thymocyte development indicated that N-CoR was a required component of short-term active repression by nuclear receptors and MAD and of a subset of long-term repression events mediated by REST/NRSF. Unexpectedly, N-CoR and a specific deacetylase were also required for transcriptional activation of one class of retinoic acid response element. Together, these findings suggest that specific combinations of corepressors and histone deacetylases mediate the gene-specific actions of DNA-bound repressors in development of multiple organ systems.


Subject(s)
Gene Expression Regulation, Developmental , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription, Genetic/physiology , Animals , Diencephalon/embryology , Erythropoiesis/physiology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Gene Deletion , Hematocrit , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Co-Repressor 1 , T-Lymphocytes/cytology , Thymus Gland/cytology , Thymus Gland/embryology , Yolk Sac/blood supply , Yolk Sac/physiology
9.
Proc Natl Acad Sci U S A ; 97(8): 4363-8, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10760302

ABSTRACT

Members of the nuclear receptor superfamily are thought to activate transcription by recruitment of one or more recently identified coactivator complexes. Here we demonstrate that both peroxisome proliferator-activated receptor binding protein (PBP) and steroid receptor coactivator-1 (SRC-1) are required for ligand-dependent transcription of transiently transfected and chromosomally integrated reporter genes by the estrogen receptor (ER) and retinoic acid receptor (RAR). To examine ligand-dependent interactions between nuclear receptors and specific coactivators in living cells, these proteins were tagged with cyan (CFP) and yellow (YFP) mutants of the green fluorescent protein. Fluorescence resonance energy transfer (FRET) from the CFP to the YFP indicated interaction between the receptor and coactivator. CFP fusions to RAR or its ligand-binding domain exhibited rapid ligand-dependent FRET to YFP-tagged nuclear receptor interaction domains of the coactivators SRC-1 and PBP. The ER-ligand-binding domain, unlike RAR, also exhibited some basal interaction with coactivators in unstimulated cells that was abolished by the receptor antagonists tamoxifen or ICI182,780. Inhibition of FRET by tamoxifen but not ICI182,780 could be reversed by estradiol, whereas estradiol-enhanced FRET could not be inhibited by either antagonist, indicating that ligand effects can show varying degrees of hysteresis. These findings suggest that ligand-dependent transcriptional activities of the RAR and ER require concurrent or sequential recruitment of SRC-1 and PBP-containing coactivator complexes.


Subject(s)
Carrier Proteins/metabolism , Receptors, Estrogen/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Cell Nucleus/metabolism , Energy Transfer , Fluorescence , Green Fluorescent Proteins , HeLa Cells , Histone Acetyltransferases , Humans , Ligands , Luminescent Proteins/metabolism , Mediator Complex Subunit 1 , Molecular Sequence Data , Nuclear Receptor Coactivator 1 , Protein Binding
10.
Mol Cell Biol ; 20(9): 3079-85, 2000 May.
Article in English | MEDLINE | ID: mdl-10757792

ABSTRACT

We have recently identified the Raf kinase inhibitor protein (RKIP) as a physiological endogenous inhibitor of the Raf-1/MEK/extracellular signal-regulated kinase (ERK) pathway. RKIP interfered with MEK phosphorylation and activation by Raf-1, resulting in the suppression of both Raf-1-induced transformation and AP-1-dependent transcription. Here we report the molecular mechanism of RKIP's inhibitory function. RKIP can form ternary complexes with Raf-1, MEK, and ERK. However, whereas MEK and ERK can simultaneously associate with RKIP, Raf-1 binding to RKIP and that of MEK are mutually exclusive. RKIP is able to dissociate a Raf-1-MEK complex and behaves as a competitive inhibitor of MEK phosphorylation. Mapping of the binding domains showed that MEK and Raf-1 bind to overlapping sites in RKIP, whereas MEK and RKIP associate with different domains in Raf-1, and Raf-1 and RKIP bind to different sites in MEK. Both the Raf-1 and the MEK binding sites in RKIP need to be destroyed in order to relieve RKIP-mediated suppression of the Raf-1/MEK/ERK pathway, indicating that binding of either Raf-1 or MEK is sufficient for inhibition. The properties of RKIP reveal the specific sequestration of interacting components as a novel motif in the cell's repertoire for the regulation of signaling pathways.


Subject(s)
Androgen-Binding Protein , Carrier Proteins/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Signal Transduction , Alleles , Carrier Proteins/genetics , Genes, Reporter , Glutathione Transferase/metabolism , Models, Biological , Phospholipid Transfer Proteins , Plasmids , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Recombinant Fusion Proteins/metabolism , Two-Hybrid System Techniques
11.
Curr Pharm Des ; 6(1): 59-98, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10637372

ABSTRACT

SH2 domains are discrete structural motifs common to a variety of critical intracellular signaling proteins. Inhibitors of specific SH2 domains have become important therapeutic targets in the treatment and/or prevention of restenosis, cancers (including small cell lung), cardiovascular disease, osteoporosis, apoptosis among others. Considering the social and economic impact of these diseases significant attention has been focused on the development of potent and selective inhibitors of specific SH2 domains. In particular, considerable research has been performed on Src, PI 3-kinase, Grb2 and more recently, Lck. In this review, we will focus on progress in the development of inhibitors for these specific SH2 domains and evaluate potential future targets.


Subject(s)
Enzyme Inhibitors/pharmacology , src Homology Domains/drug effects , Amino Acid Sequence , Animals , Drug Design , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/chemistry , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry
12.
Nature ; 401(6749): 173-7, 1999 Sep 09.
Article in English | MEDLINE | ID: mdl-10490027

ABSTRACT

Raf-1 phosphorylates and activates MEK-1, a kinase that activates the extracellular signal regulated kinases (ERK). This kinase cascade controls the proliferation and differentiation of different cell types. Here we describe a Raf-1-interacting protein, isolated using a yeast two-hybrid screen. This protein inhibits the phosphorylation and activation of MEK by Raf-1 and is designated RKIP (Raf kinase inhibitor protein). In vitro, RKIP binds to Raf-1, MEK and ERK, but not to Ras. RKIP co-immunoprecipitates with Raf-1 and MEK from cell lysates and colocalizes with Raf-1 when examined by confocal microscopy. RKIP is not a substrate for Raf-1 or MEK, but competitively disrupts the interaction between these kinases. RKIP overexpression interferes with the activation of MEK and ERK, induction of AP-1-dependent reporter genes and transformation elicited by an oncogenically activated Raf-1 kinase. Downregulation of endogenous RKIP by expression of antisense RNA or antibody microinjection induces the activation of MEK-, ERK- and AP-1-dependent transcription. RKIP represents a new class of protein-kinase-inhibitor protein that regulates the activity of the Raf/MEK/ERK module.


Subject(s)
Androgen-Binding Protein , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Carrier Proteins/metabolism , MAP Kinase Kinase Kinase 1 , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Signal Transduction/drug effects , 3T3 Cells , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carrier Proteins/isolation & purification , Cell Transformation, Neoplastic , Cloning, Molecular , Enzyme Activation , Enzyme Inhibitors/metabolism , Gene Expression Regulation , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mice , Phosphatidylethanolamine Binding Protein , Phospholipid Transfer Proteins , Prostatein , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Secretoglobins , Transcription Factor AP-1/metabolism , Uteroglobin
13.
Mol Cell Biol ; 19(9): 6367-78, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10454583

ABSTRACT

Nuclear factor-kappaB (NF-kappaB) plays a role in the transcriptional regulation of genes involved in inflammation and cell survival. In this report we demonstrate that NF-kappaB recruits a coactivator complex that has striking similarities to that recruited by nuclear receptors. Inactivation of either cyclic AMP response element binding protein (CREB)-binding protein (CBP), members of the p160 family of coactivators, or the CBP-associated factor (p/CAF) by nuclear antibody microinjection prevents NF-kappaB-dependent transactivation. Like nuclear receptor-dependent gene expression, NF-kappaB-dependent gene expression requires specific LXXLL motifs in one of the p160 family members, and enhancement of NF-kappaB activity requires the histone acetyltransferase (HAT) activity of p/CAF but not that of CBP. This coactivator complex is differentially recruited by members of the Rel family. The p50 homodimer fails to recruit coactivators, although the p50-p65 heterodimeric form of the transcription factor assembles the integrator complex. These findings provide new mechanistic insights into how this family of dimeric transcription factors has a differential effect on gene expression.


Subject(s)
NF-kappa B/metabolism , Saccharomyces cerevisiae Proteins , Transcriptional Activation , Acetyltransferases/genetics , Acetyltransferases/metabolism , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Histone Acetyltransferases , NF-kappa B/genetics , Nuclear Receptor Coactivator 1 , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , p300-CBP Transcription Factors
14.
Nat Genet ; 22(4): 394-9, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10431247

ABSTRACT

LIM domains are required for both inhibitory effects on LIM homeodomain transcription factors and synergistic transcriptional activation events. The inhibitory actions of the LIM domain can often be overcome by the LIM co-regulator known as CLIM2, LDB1 and NLI (referred to hereafter as CLIM2; refs 2-4). The association of the CLIM cofactors with LIM domains does not, however, improve the DNA-binding ability of LIM homeodomain proteins, suggesting the action of a LIM-associated inhibitor factor. Here we present evidence that LIM domains are capable of binding a novel RING-H2 zinc-finger protein, Rlim (for RING finger LIM domain-binding protein), which acts as a negative co-regulator via the recruitment of the Sin3A/histone deacetylase corepressor complex. A corepressor function of RLIM is also suggested by in vivo studies of chick wing development. Overexpression of the gene Rnf12, encoding Rlim, results in phenotypes similar to those observed after inhibition of the LIM homeodomain factor LHX2, which is required for the formation of distal structures along the proximodistal axis, or by overexpression of dominant-negative CLIM1. We conclude that Rlim is a novel corepressor that recruits histone deacetylase-containing complexes to the LIM domain.


Subject(s)
Histone Deacetylases/metabolism , Homeodomain Proteins/antagonists & inhibitors , Repressor Proteins/physiology , Transcription Factors/antagonists & inhibitors , Amino Acid Sequence , Animals , COS Cells , Chick Embryo , Extremities/anatomy & histology , Extremities/embryology , Gene Expression Regulation, Developmental , Mice , Molecular Sequence Data , Repressor Proteins/analysis , Sequence Homology, Amino Acid , Time Factors , Tissue Distribution , Transfection , Ubiquitin-Protein Ligases
15.
Nat Med ; 5(6): 635-42, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10371501

ABSTRACT

The HIV-1 Rev protein facilitates the nuclear export of mRNA containing the Rev response element (RRE) through binding to the export receptor CRM-1. Here we show that a cellular nuclear protein, Sam68 (Src-associated protein in mitosis), specifically interacts with RRE and can partially substitute for as well as synergize with Rev in RRE-mediated gene expression and virus replication. Differential sensitivity to leptomycin B, an inhibitor of CRM-1, indicates that the export pathways mediated by Rev and Sam68 are distinct. C-terminally deleted mutants of Sam68 inhibited the transactivation of RRE-mediated expression by both wild-type Sam68 and Rev. They were retained in the cytoplasm and impeded the nuclear localization of Rev in co-expressed cells. These mutants also inhibited wild-type HIV-1 replication to the same extent as the RevM10 mutant, and may be useful as anti-viral agents in the treatment of AIDS.


Subject(s)
Gene Products, rev , HIV-1/physiology , Karyopherins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear , Virus Replication/genetics , Adaptor Proteins, Signal Transducing , Antibodies/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cells, Cultured/virology , Chloramphenicol O-Acetyltransferase/genetics , Cytoplasm/metabolism , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Fatty Acids, Unsaturated/pharmacology , Gene Expression Regulation, Viral , Genes, Dominant , Genes, Reporter , HeLa Cells/virology , Humans , Kinetin , Mutation , Purines/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , Response Elements , rev Gene Products, Human Immunodeficiency Virus , Exportin 1 Protein
16.
Proc Natl Acad Sci U S A ; 96(7): 3652-7, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097092

ABSTRACT

CREB-binding proteins (CBP) and p300 are essential transcriptional coactivators for a large number of regulated DNA-binding transcription factors, including CREB, nuclear receptors, and STATs. CBP and p300 function in part by mediating the assembly of multiprotein complexes that contain additional cofactors such as p300/CBP interacting protein (p/CIP), a member of the p160/SRC family of coactivators, and the p300/CBP associated factor p/CAF. In addition to serving as molecular scaffolds, CBP and p300 each possess intrinsic acetyltransferase activities that are required for their function as coactivators. Here we report that the adenovirus E1A protein inhibits the acetyltransferase activity of CBP on binding to the C/H3 domain, whereas binding of CREB, or a CREB/E1A fusion protein to the KIX domain, fails to inhibit CBP acetyltransferase activity. Surprisingly, p/CIP can either inhibit or stimulate CBP acetyltransferase activity depending on the specific substrate evaluated and the functional domains present in the p/CIP protein. While the CBP interaction domain of p/CIP inhibits acetylation of histones H3, H4, or high mobility group by CBP, it enhances acetylation of other substrates, such as Pit-1. These observations suggest that the acetyltransferase activities of CBP/p300 and p/CAF can be differentially modulated by factors binding to distinct regions of CBP/p300. Because these interactions are likely to result in differential effects on the coactivator functions of CBP/p300 for different classes of transcription factors, regulation of CBP/p300 acetyltransferase activity may represent a mechanism for integration of diverse signaling pathways.


Subject(s)
Acetyltransferases/metabolism , Adenovirus E1A Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Acetylation , Animals , Binding Sites , CREB-Binding Protein , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Fibroblasts , Histones/metabolism , Kinetics , Rats , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/metabolism , Substrate Specificity , Transfection
17.
J Biol Chem ; 274(5): 3001-8, 1999 Jan 29.
Article in English | MEDLINE | ID: mdl-9915838

ABSTRACT

Crk is a member of a family of adapter proteins predominantly composed of Src homology 2 and 3 domains, whose role in signaling pathways is presently unclear. Using an in situ electroporation system which permits the introduction of glutathione S-transferase (GST) fusion proteins into cells, we found that c-CrkII bound to p130(cas), but not to paxillin in serum-starved rat-1 fibroblasts overexpressing the human insulin receptor (HIRc cells) in vivo. 17 nM insulin stimulation dissociated the binding of c-CrkII to p130(cas), whereas 13 nM insulin-like growth factor-I, 16 nM epidermal growth factor (EGF), and 10% serum each showed little or no effect. We found that stress fiber formation is consistent with a change in the p130(cas).c-CrkII interactions before and after growth factor stimulation. Microinjection of either GST-Crk-SH2 or -Crk-(N)SH3 domains, or anti-Crk antibody each inhibited stress fiber formation before and after insulin-like growth factor-I, EGF, and serum stimulation. Insulin stimulation by itself caused stress fiber breakdown and there was no additive effect of microinjection. Microinjection of anti-p130(cas) antibody also blocked stress fiber formation in quiescent cells. Microinjection of the Crk-inhibitory reagents also inhibited DNA synthesis after insulin-like growth factor-I, EGF, and serum stimulation, but not after insulin. These data suggest that the complex containing p130(cas).c-CrkII may play a crucial role in actin cytoskeleton organization and in anchorage-dependent DNA synthesis.


Subject(s)
Actins/physiology , Cytoskeleton/physiology , Protein Kinases/physiology , Ubiquitin-Protein Ligases , src Homology Domains , Cells, Cultured , Cytoskeleton/drug effects , DNA Replication , Electroporation , Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/pharmacology , Humans , Insulin/administration & dosage , Insulin/pharmacology , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Microinjections , Mitosis , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-cbl , Proto-Oncogene Proteins c-crk , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , Tyrosine/metabolism
18.
Proc Natl Acad Sci U S A ; 96(2): 709-14, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892698

ABSTRACT

Retroviruses must bypass the tight coupling of splicing and nuclear export of mRNA in their replication cycle because unspliced genomic RNA and incompletely spliced mRNA must be exported to the cytoplasm for packaging or translation. This process is mediated by a cis-acting constitutive transport element (CTE) for simple retroviruses and by the trans-acting viral protein Rev in concert with its response element (RRE) for complex retroviruses (e.g., HIV). Recently, we identified RNA helicase A (RHA) as a potential cellular cofactor for CTE. Here, we report that RHA also plays a role in Rev/RRE-mediated gene expression and HIV replication. RHA binds weakly to HIV-1 RRE independently of Rev. Overexpression of RHA, but not of an RHA mutant lacking helicase activity, increased both Rev/RRE- and CTE-dependent gene expression and the levels of unspliced HIV mRNA. Microinjection of antibodies to RHA into nuclei dramatically inhibited both CTE- and Rev-dependent gene expression in human cells. Exogenous RHA cDNA, but not the mutant RHA, rescued this inhibition. We propose that RHA is required to release both CTE- and RRE-containing mRNA from spliceosomes before completion of splicing, thus freeing them for nuclear export.


Subject(s)
HIV-1/enzymology , RNA Helicases/metabolism , RNA Processing, Post-Transcriptional/genetics , Antibodies/pharmacology , DNA, Complementary/genetics , Gene Expression Regulation, Viral/genetics , Gene Products, rev/genetics , Genes, env/genetics , HIV Core Protein p24/genetics , HeLa Cells , Humans , Immunohistochemistry , Microinjections , Mutation/genetics , Nuclear Proteins/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Virus Replication/genetics , rev Gene Products, Human Immunodeficiency Virus
19.
Genes Dev ; 13(24): 3198-208, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10617569

ABSTRACT

Retinoic acid and thyroid hormone receptors can act alternatively as ligand-independent repressors or ligand-dependent activators, based on an exchange of N-CoR or SMRT-containing corepressor complexes for coactivator complexes in response to ligands. We provide evidence that the molecular basis of N-CoR recruitment is similar to that of coactivator recruitment, involving cooperative binding of two helical interaction motifs within the N-CoR carboxyl terminus to both subunits of a RAR-RXR heterodimer. The N-CoR and SMRT nuclear receptor interaction motifs exhibit a consensus sequence of LXX I/H I XXX I/L, representing an extended helix compared to the coactivator LXXLL helix, which is able to interact with specific residues in the same receptor pocket required for coactivator binding. We propose a model in which discrimination of the different lengths of the coactivator and corepressor interaction helices by the nuclear receptor AF2 motif provides the molecular basis for the exchange of coactivators for corepressors, with ligand-dependent formation of the charge clamp that stabilizes LXXLL binding sterically inhibiting interaction of the extended corepressor helix.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Consensus Sequence , Conserved Sequence , Dimerization , HeLa Cells , Humans , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Receptor Co-Repressor 1 , Protein Structure, Secondary , Rats , Receptors, Retinoic Acid/chemistry , Receptors, Thyroid Hormone/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Retinoid X Receptors , Sequence Alignment , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Transcription Factors/metabolism
20.
Genes Dev ; 12(21): 3357-68, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9808623

ABSTRACT

Ligand-dependent activation of gene transcription by nuclear receptors is dependent on the recruitment of coactivators, including a family of related NCoA/SRC factors, via a region containing three helical domains sharing an LXXLL core consensus sequence, referred to as LXDs. In this manuscript, we report receptor-specific differential utilization of LXXLL-containing motifs of the NCoA-1/SRC-1 coactivator. Whereas a single LXD is sufficient for activation by the estrogen receptor, different combinations of two, appropriately spaced, LXDs are required for actions of the thyroid hormone, retinoic acid, peroxisome proliferator-activated, or progesterone receptors. The specificity of LXD usage in the cell appears to be dictated, at least in part, by specific amino acids carboxy-terminal to the core LXXLL motif that may make differential contacts with helices 1 and 3 (or 3') in receptor ligand-binding domains. Intriguingly, distinct carboxy-terminal amino acids are required for PPARgamma activation in response to different ligands. Related LXXLL-containing motifs in NCoA-1/SRC-1 are also required for a functional interaction with CBP, potentially interacting with a hydrophobic binding pocket. Together, these data suggest that the LXXLL-containing motifs have evolved to serve overlapping roles that are likely to permit both receptor-specific and ligand-specific assembly of a coactivator complex, and that these recognition motifs underlie the recruitment of coactivator complexes required for nuclear receptor function.


Subject(s)
Peptide Fragments/chemistry , Receptors, Cytoplasmic and Nuclear/chemistry , Transcription Factors/chemistry , Transcriptional Activation/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Fibroblasts/cytology , Gene Expression Regulation , Histone Acetyltransferases , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/physiology , Nuclear Receptor Coactivator 1 , Peptide Fragments/genetics , Peptide Fragments/physiology , Protein Structure, Secondary , Rats , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Sequence Alignment , Trans-Activators/physiology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...