Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Oncogene ; 35(26): 3410-8, 2016 06 30.
Article in English | MEDLINE | ID: mdl-26477307

ABSTRACT

Aberrant activation of Wnt/ß-catenin pathway contributes to colorectal cancer (CRC) progression. However, little is known about regulatory mechanisms of the ß-catenin activity in cancer progression. Here we investigated the role of DBC1, which was recently reported as a negative regulator of SIRT1 and a transcriptional coactivator, in the regulation of Wnt/ß-catenin signaling. We identified the genome-wide targets of DBC1 and found that loss of DBC1 inhibits the expression of ß-catenin target genes including PROX1, a transcription factor linked to CRC progression. Mechanistically, DBC1 stabilizes LEF1-ß-catenin interaction by inhibiting SIRT1-mediated ß-catenin deacetylation, thereby enhancing LEF1-ß-catenin complex formation and long-range chromatin looping at the PROX1 locus. Furthermore, DBC1 is also required for the transcriptional activity of PROX1, suggesting that DBC1 has a dual function in regulating ß-catenin-PROX1 signaling axis: as a coactivator for both ß-catenin and PROX1. Importantly, loss of DBC1 inhibited growth and tumorigenic potential of colon cancer cells, and DBC1 expression correlated with shorter relapse-free survival in patients with advanced CRC. Our results firmly establish DBC1 as a critical positive regulator of ß-catenin-PROX1 signaling axis and a key factor in ß-catenin-PROX1-mediated CRC progression.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Colorectal Neoplasms/genetics , Homeodomain Proteins/genetics , Signal Transduction/genetics , beta Catenin/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , HT29 Cells , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Mice, Inbred BALB C , Mice, Nude , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Suppressor Proteins , beta Catenin/metabolism
2.
Oncogene ; 32(35): 4160-8, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-22986521

ABSTRACT

Estrogen receptor α (ERα) has critical roles in the development and progression of breast cancer, and the coiled-coil co-activator (CoCoA) is an important ERα co-activator for estrogen-induced gene expression. The small ubiquitin-like modifier (SUMO) pathway is hyperactivated in breast cancer, but the mechanism by which SUMOylation regulates ERα-mediated transcription remains poorly understood. Here, we identified ZFP282 as a CoCoA-binding protein. ZFP282 associates directly with ERα and cooperates synergistically with CoCoA to enhance ERα function. ZFP282 is required for estrogen-induced expression of ERα target genes and estrogen-dependent breast cancer cell growth and tumorigenesis. In addition, we found that ZFP282 is SUMOylated and that SUMOylation positively regulates the co-activator activity of ZFP282 by increasing its binding affinity to ERα and CoCoA, and consequently increasing recruitment of ZFP282-CoCoA complex to the promoter of ERα target genes. These findings reveal essential roles for ZFP282 and its SUMOylation in estrogen signaling and breast tumorigenesis.


Subject(s)
Breast Neoplasms/etiology , Estrogen Receptor alpha/physiology , Signal Transduction/physiology , Sumoylation , Zinc Fingers , Cell Proliferation , Cell Transformation, Neoplastic , Female , Human T-lymphotropic virus 1/genetics , Humans , Terminal Repeat Sequences , Transcription, Genetic
3.
Oncogene ; 31(39): 4290-301, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22249259

ABSTRACT

Linker histone H1.2 has been shown to suppress p53-dependent transcription through the modulation of chromatin remodeling; however, little is known about the mechanisms governing the antagonistic effects of H1.2 in DNA damage response. Here, we show that the repressive action of H1.2 on p53 function is negatively regulated via acetylation of p53 C-terminal regulatory domain and phosphorylation of H1.2 C-terminal tail. p53 acetylation by p300 impairs the interaction of p53 with H1.2 and triggers a rapid activation of p53-dependent transcription. Similarly, DNA-PK-mediated phosphorylation of H1.2 at T146 enhances p53 transcriptional activity by impeding H1.2 binding to p53 and thereby attenuating its suppressive effects on p53 transactivation. Consistent with these findings, point mutations mimicking modification states of H1.2 and p53 lead to a significant increase in p53-induced apoptosis. These data suggest that p53 acetylation-H1.2 phosphorylation cascade serves as a unique mechanism for triggering p53-dependent DNA damage response pathways.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Acetylation , Apoptosis/genetics , Cell Line , DNA Damage , DNA Repair , Histones/genetics , Humans , Phosphorylation , Point Mutation , Tumor Suppressor Protein p53/genetics
4.
J Mol Endocrinol ; 34(1): 107-18, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15691881

ABSTRACT

Androgen-independent prostate cancer is a lethal form of the disease that is marked by metastasis and rapid proliferation in its final stages. As no effective therapy for this aggressive tumor currently exists, it is imperative to elucidate and target the mechanisms involved in the progression to androgen independence. Accumulating evidence indicates that aberrant activation of androgen receptor (AR) via signal transduction pathways, AR gene mutation and/or amplification, and/or coregulator alterations may contribute to the progression of prostate cancer. In the present study, the effects of protein kinase A (PKA) signaling and its downstream factors on AR activity at the prostate-specific antigen (PSA) gene were tested. Activation of PKA by forskolin resulted in enhanced androgen-induced expression of the PSA gene, an effect that was blocked by the AR antagonist, bicalutamide. Interestingly, when either p300 or CBP was overexpressed, PKA activation was sufficient to stimulate PSA promoter-driven transcription in the absence of androgen, which was not inhibited by bicalutamide. PKA activation did not significantly alter AR protein levels but significantly increased the phosphorylated form of its downstream effector, cAMP responsive element-binding protein (CREB) in the presence of androgen. Furthermore, chromatin immunoprecipitation showed that the combination of androgen and forskolin increased phosphorylated CREB occupancy, which was accompanied by histone acetylation, at the putative cAMP responsive element located in the 5' upstream regulatory region of the PSA gene. Remarkably, mammalian two-hybrid assay indicated that p300/CBP may bridge the interaction between AR and CREB, suggesting a novel enhanceosomal cooperation. These results demonstrate an intriguing interplay between a signal transduction pathway, coactivator overexpression and AR signaling as a possible combined mechanism of progression to androgen-independent prostate cancer.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Prostate-Specific Antigen/genetics , Receptors, Androgen/metabolism , Transcription, Genetic/physiology , Acetylation , Colforsin/pharmacology , Histones/metabolism , Humans , Male , Phosphorylation , Prostate-Specific Antigen/metabolism , Transcription, Genetic/drug effects
5.
Curr Biol ; 11(24): 1981-5, 2001 Dec 11.
Article in English | MEDLINE | ID: mdl-11747826

ABSTRACT

Activation of gene transcription involves chromatin remodeling by coactivator proteins that are recruited by DNA-bound transcription factors. Local modification of chromatin structure at specific gene promoters by ATP-dependent processes and by posttranslational modifications of histone N-terminal tails provides access to RNA polymerase II and its accompanying transcription initiation complex. While the roles of lysine acetylation, serine phosphorylation, and lysine methylation of histones in chromatin remodeling are beginning to emerge, low levels of arginine methylation of histones have only recently been documented, and its physiological role is unknown. The coactivator CARM1 methylates histone H3 at Arg17 and Arg26 in vitro and cooperates synergistically with p160-type coactivators (e.g., GRIP1, SRC-1, ACTR) and coactivators with histone acetyltransferase activity (e.g., p300, CBP) to enhance gene activation by steroid and nuclear hormone receptors (NR) in transient transfection assays. In the current study, CARM1 cooperated with GRIP1 to enhance steroid hormone-dependent activation of stably integrated mouse mammary tumor virus (MMTV) promoters, and this coactivator function required the methyltransferase activity of CARM1. Chromatin immunoprecipitation assays and immunofluorescence studies indicated that CARM1 and the CARM1-methylated form of histone H3 specifically associated with a large tandem array of MMTV promoters in a hormone-dependent manner. Thus, arginine-specific histone methylation by CARM1 is an important part of the transcriptional activation process.


Subject(s)
Arginine/metabolism , Histones/metabolism , Hormones/physiology , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases/physiology , Steroids/physiology , Acetylation , Fluorescent Antibody Technique , Histones/chemistry , Lysine/metabolism , Mammary Tumor Virus, Mouse/genetics , Methylation , Phosphorylation , Precipitin Tests , Serine/metabolism
6.
Curr Biol ; 11(12): 996-1000, 2001 Jun 26.
Article in English | MEDLINE | ID: mdl-11448779

ABSTRACT

Posttranslational modifications of histone amino termini play an important role in modulating chromatin structure and function. Lysine methylation of histones has been well documented, and recently this modification has been linked to cellular processes involving gene transcription and heterochromatin assembly. However, the existence of arginine methylation on histones has remained unclear. Recent discoveries of protein arginine methyltransferases, CARM1 and PRMT1, as transcriptional coactivators for nuclear receptors suggest that histones may be physiological targets of these enzymes as part of a poorly defined transcriptional activation pathway. Here we show by using mass spectrometry that histone H4, isolated from asynchronously growing human 293T cells, is methylated at arginine 3 (Arg-3) in vivo. In support, a novel antibody directed against histone H4 methylated at Arg-3 independently demonstrates the in vivo occurrence of this modification and reveals that H4 Arg-3 methylation is highly conserved throughout eukaryotes. Finally, we show that PRMT1 is the major, if not exclusive, H4 Arg-3 methyltransfase in human 293T cells. These findings suggest a role for arginine methylation of histones in the transcription process.


Subject(s)
Histones/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Amino Acid Motifs , Animals , Arginine/metabolism , Cell Line , Genes, Reporter , Humans , Immunoblotting , Methylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
7.
Oncogene ; 20(24): 3014-20, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11420716

ABSTRACT

Recent findings suggest that lysine and arginine-specific methylation of histones may cooperate with other types of post-translational histone modification to regulate chromatin structure and gene transcription. Proteins that methylate histones on arginine residues can collaborate with other coactivators to enhance the activity of specific transcriptional activators such as nuclear receptors. Lysine methylation of histones is associated with transcriptionally active nuclei, regulates other types of histone modifications, and is necessary for proper mitotic cell divisions. The fact that some transcription factors and proteins involved in RNA processing can also be methylated suggests that protein methylation may also contribute in other ways to regulation of transcription and post-transcriptional steps in gene regulation. In future work, it will be important to develop methods for evaluating the precise roles of protein methylation in the regulation of native genes in physiological settings, e.g. by using chromatin immunoprecipitation assays, differentiating cell culture systems, and genetically altered cells and animals. It will also be important to isolate additional protein methyltransferases by molecular cloning and to characterize new methyltransferase substrates, the regulation of methyltransferase activities, and the roles of new methyltransferases and substrates.


Subject(s)
Chromatin/genetics , Proteins/metabolism , Transcription, Genetic , Animals , Chromatin/metabolism , Gene Expression Regulation , Histones/metabolism , Humans , Methylation
8.
Biochemistry ; 40(19): 5747-56, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11341840

ABSTRACT

The preferential in vitro methylation of histone H3 by coactivator-associated arginine methyltransferase 1 (CARM1) has been proposed as a basis for its ability to enhance gene transcription [Chen, D., et al. (1999) Science 284, 2174-2177]. To further evaluate the significance of H3 methylation, we studied the kinetics and site specificity of its modification by CARM1. Affinity-purified CARM1 methylated recombinant chick H3, which is free of posttranslational modifications, and calf thymus H3, which is heterogeneous with regard to preexisting modifications, equally well, exhibiting a V(max) of 4500 pmol min(-1) (mg of enzyme)(-1) and an apparent K(m) for H3 of < or = 0.2 microM. The catalytic efficiency (k(cat)/K(m)) of CARM1 toward H3 was at least 1000 times that toward R1 (GGFGGRGGFGG-amide), a highly effective substrate for protein arginine methyltransferase 1. Peptide mapping of 3H-methyl-labeled H3 indicated methylation at Arg-2, Arg-17, and Arg-26 in the N-terminal region and at one or more of four arginines (128/129/131/134) at the C-terminus. Two of the N-terminal sites, Arg-17 and Arg-26, occur in the sequence KAXRK and appear to be more efficiently methylated than Arg-2. CARM1 catalyzed formation of N(G),N(G)-dimethylarginine (asymmetric) but little or no N(G),N'(G)-dimethylarginine (symmetric) and no form of methyllysine. Amino acid analysis of untreated calf thymus H3 revealed that 3.7% of the molecules naturally contain asymmetric dimethylarginine and/or monomethylarginine. Our findings support the hypothesis that methylation of H3 may be involved in the mechanism of transcriptional coactivation by CARM1 of genes whose expression is under the control of nuclear receptors.


Subject(s)
Histones/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Catalysis , Cattle , Chickens , Chromatography, High Pressure Liquid , Enzyme Activation , Histones/genetics , Kinetics , Methylation , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Substrate Specificity/genetics , omega-N-Methylarginine/metabolism
9.
Oncogene ; 20(17): 2134-43, 2001 Apr 19.
Article in English | MEDLINE | ID: mdl-11360197

ABSTRACT

A recently discovered potential tumor suppressor protein, Zac1, was previously shown to promote cell cycle arrest and apoptosis, and to act as a positive or negative transcriptional cofactor for nuclear receptors. Since these activities are common to Zac1 and p53, we tested for a functional interaction between these two proteins by investigating possible effects of Zac1 on the transcriptional activator function of p53. Zac1 specifically enhanced the activity of p53-responsive promoters in cells expressing wild type p53. The same promoters were not activated by Zac1 in cells lacking functional p53, but the Zac1 effect was restored by co-expression of p53. Zac1 bound to p53 and enhanced the activity of p53 or its N-terminal transcriptional activation domain fused to the DNA binding domain of Gal4. These results indicate that Zac1 served as a transcriptional coactivator for p53. The enhancement of p53 activity by Zac1 was much more dramatic in HeLa cells than in other cell lines tested. HeLa cells express human papillomavirus type 18 E6 protein which inactivates and causes the degradation of p53. Physical and functional interactions observed between Zac1 and E6 protein indicated that the dramatic activity of Zac1 in HeLa cells was due not only to Zac1's coactivator effect on p53, but also to the ability of Zac1 to reverse E6 inhibition of p53.


Subject(s)
Cell Cycle Proteins/physiology , DNA-Binding Proteins , Genes, Tumor Suppressor , Trans-Activators/physiology , Transcription Factors , Tumor Suppressor Protein p53/physiology , Animals , COS Cells , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Neoplastic , Genes, Reporter , Genes, p53 , HeLa Cells , Humans , Oncogene Proteins, Viral/physiology , Response Elements , Trans-Activators/metabolism , Transcriptional Activation/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins
10.
J Biol Chem ; 276(25): 22177-82, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11301320

ABSTRACT

Promoter-bound steroid receptors activate gene expression by recruiting members of the p160 family of coactivators. Many steroid receptors, most notably the progesterone and estrogen receptors, are regulated both by cognate hormone and independently by growth factors. Here we show that epidermal growth factor regulates the activities of the p160 GRIP1 through the extracellular signal-regulated kinase (ERK) family of mitogen-activated protein kinases. ERKs phosphorylate GRIP1 at a specific site, Ser-736, the integrity of which is required for full growth factor induction of GRIP1 transcriptional activation and coactivator function. We propose that growth factors signal to nuclear receptors in part by targeting the p160 coactivators.


Subject(s)
Epidermal Growth Factor/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Steroid/metabolism , Signal Transduction , Transcription Factors/metabolism , HeLa Cells , Humans , Nuclear Receptor Coactivator 2 , Phosphorylation
11.
Mol Endocrinol ; 15(4): 485-500, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11266502

ABSTRACT

The glucocorticoid receptor interacting protein-1 (GRIP1) is a member of the steroid receptor coactivator (SRC) family of transcriptional regulators. Green fluorescent protein (GFP) fusions were made to full-length GRIP1, and a series of GRIP1 mutants lacking the defined regulatory regions and the intracellular distribution of these proteins was studied in HeLa cells. The distribution of GRIP1 was complex, ranging from diffuse nucleoplasmic to discrete intranuclear foci. Formation of these foci was dependent on the C-terminal region of GRIP1, which contains the two characterized transcriptional activation domains, AD1 and AD2. A subpopulation of GRIP1 foci associate with ND10s, small nuclear bodies that contain several proteins including PML, SP100, DAXX, and CREB-binding protein (CBP). Association with the ND10s is dependent on the AD1 of GRIP1, a region of the protein previously described as a CBP-interacting domain. The GRIP1 foci are enriched in components of the 26S proteasome, including the core 20S proteasome, PA28alpha, and ubiquitin. In addition, the irreversible proteasome inhibitor lactacystin induced an increase in the total fluorescence intensity of the GFP-GRIP1 expressing cells, demonstrating that GRIP1 is degraded by the proteasome. These findings suggest the intriguing possibility that degradation of GRIP1 by the 26S proteasome may be a key component of its regulation.


Subject(s)
Acetylcysteine/analogs & derivatives , Antigens, Nuclear , Cell Nucleus Structures/metabolism , Intracellular Signaling Peptides and Proteins , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex , Transcription Factors/metabolism , Acetylcysteine/pharmacology , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Autoantigens/metabolism , Base Sequence , Binding Sites , CREB-Binding Protein , Carrier Proteins/metabolism , Co-Repressor Proteins , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Molecular Chaperones , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2 , Nuclear Receptor Coactivator 3 , Peptide Hydrolases/drug effects , Promyelocytic Leukemia Protein , Protease Inhibitors/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Trans-Activators/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins , Ubiquitins/metabolism
12.
J Biol Chem ; 276(2): 1089-98, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11050077

ABSTRACT

Nuclear receptors (NRs) activate gene transcription by binding to specific enhancer elements and recruiting coactivators of the p160 family to promoters of target genes. The p160 coactivators in turn enhance transcription by recruiting secondary coactivators, including histone acetyltransferases such as CREB-binding protein (CBP) and p300/CBP-associated factor (p/CAF), as well as the recently identified protein methyltransferase, coactivator-associated arginine methyltransferase 1 (CARM1). In the current study, protein arginine methyltransferase 1 (PRMT1), another arginine-specific protein methyltransferase that shares a region of high homology with CARM1, was also found to act as a coactivator for NRs. PRMT1, like CARM1, bound to the C-terminal AD2 activation domain of p160 coactivators and thereby enhanced the activity of NRs in transient transfection assays. The shape of the graphs of reporter gene activity versus the amounts of CARM1 or PRMT1 expression vector indicated a cooperative relationship between coactivator concentration and activity. Moreover, CARM1 and PRMT1 acted in a synergistic manner to enhance reporter gene activation by both hormone-dependent and orphan NRs. The synergy was most evident at low levels of transfected NR expression vectors, where activation of reporter genes was almost completely dependent on the presence of NR and all three exogenously supplied coactivators, i.e. GRIP1, CARM1, and PRMT1. In contrast, with the higher levels of NR expression vectors typically used in transient transfection assays, NR activity was much less dependent on the combination of coactivators, suggesting that target gene activation occurs by different mechanisms at high versus low cellular concentrations of NR. Because multiple coactivators are presumably required to mediate transcriptional activation of native genes in vivo, the low-NR conditions may provide a more physiologically relevant assay for coactivator function.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Cell Line , Enzyme Activation , Genes, Reporter , Glutathione Transferase/genetics , Kinetics , Mammals , Nuclear Receptor Coactivator 2 , Recombinant Proteins/metabolism , Transfection
13.
Cancer Res ; 60(21): 5946-9, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085509

ABSTRACT

In the present study, the role of BRCA1 in ligand-dependent androgen receptor (AR) signaling was assessed. In transfected prostate and breast cancer cell lines, BRCA1 enhanced AR-dependent transactivation of a probasin-derived reporter gene. The effects of BRCA1 were mediated through the NH2-terminal activation function (AF-1) of the receptor. Cotransfection of p160 coactivators markedly potentiated BRCA1-mediated enhancement of AR signaling. In addition, BRCA1 was shown to interact physically with both the AR and the p160 coactivator, glucocorticoid receptor interacting protein 1. These findings suggest that BRCA1 may directly modulate AR signaling and, therefore, may have implications regarding the proliferation of normal and malignant androgen-regulated tissues.


Subject(s)
Genes, BRCA1/physiology , Receptors, Androgen/physiology , Signal Transduction/physiology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA1 Protein/physiology , Breast Neoplasms/genetics , Culture Techniques , Female , Gene Expression , Genes, Reporter , Humans , Male , Nuclear Receptor Coactivator 2 , Polyglutamic Acid/pharmacology , Polyglutamic Acid/physiology , Prostatic Neoplasms/genetics , Protein Structure, Tertiary , Receptors, Androgen/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism , Transcription Factors/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology , Transfection , Tumor Cells, Cultured
14.
J Biol Chem ; 275(52): 40810-6, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11010967

ABSTRACT

Members of the p160 coactivator family (steroid receptor coactivator-1 (SRC-1), glucocorticoid receptor interacting protein 1 (GRIP1), and activator of thyroid and retinoic acid receptors (ACTR)) mediate transcriptional activation by nuclear receptors. After being recruited to the promoter by nuclear receptors, the p160 coactivator transmits the activating signal via two C-terminal activation domains, AD1 and AD2. AD1 is a binding site for the related coactivators cAMP-response element binding protein binding protein (CBP) and p300, whereas AD2 binds to another coactivator, coactivator-associated arginine methyltransferase 1 (CARM1), a protein-arginine methyltransferase. The current study explored the cooperative functional and mechanistic relationships among GRIP1, CARM1, and p300 in transient transfection assays, where they enhanced the ability of the estrogen receptor (ER) to activate transcription of a reporter gene. The coactivator functions of p300 and CARM1 depended on the co-expression of GRIP1. Simultaneous co-expression of all three coactivators caused a synergistic enhancement of ER function. Deletion of the AD1 domain of GRIP1 abolished the ability of p300 to potentiate ER activity but had no effect on CARM1-mediated stimulation. In contrast, when the AD2 domain of GRIP1 was deleted, p300 still stimulated ER function through the mutant GRIP1, but CARM1 failed to do so. Thus, both binding of p300 to AD1 and binding of CARM1 to AD2 are required for their respective coactivator functions and for their synergy. Furthermore, CARM1 and p300 function independently through different activating domains of GRIP1, and their synergy suggests that they enhance transcription by different, complementary mechanisms.


Subject(s)
Nuclear Proteins/physiology , Protein-Arginine N-Methyltransferases/physiology , Receptors, Estrogen/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Cell Line , Cells, Cultured , Histone Acetyltransferases , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2
15.
Biochem Soc Trans ; 28(4): 415-8, 2000.
Article in English | MEDLINE | ID: mdl-10961931

ABSTRACT

Nuclear hormone receptors (NRs) activate transcription by binding to specific enhancer elements associated with target genes. Transcriptional activation is accomplished with the help of complexes of co-activator proteins that bind to NRs. p160 co-activators, a family of three related 160 kDa proteins, serve as primary co-activators by binding directly to NRs and recruiting additional secondary co-activators. Some of these (CBP/p300 and p/CAF) can acetylate histones and other proteins in the transcription complex, thus helping to modify chromatin structure and form an active transcription initiation complex. We recently discovered co-activator-associated arginine methyltransferase 1 (CARM1), which binds to p160 co-activators and thereby enhances transcriptional activation by NRs on transiently transfected reporter genes. CARM1 also methylates specific arginine residues in the N-terminal tail of histone H3 in vitro. A related arginine-specific protein methyltransferase, PRMT1, also binds p160 co-activators and enhances NR function. PRMT1 methylates histone H4 in vitro. The enhancement of NR function by CARM1, PRMT1 and p300 depends on their interactions with p160 co-activators. In the presence of p160 co-activators, some pairs of these three secondary co-activators provide a highly synergistic enhancement of NR function on transiently transfected reporter genes. We have also observed an enhancement of NR function on stably integrated reporter genes by these co-activators. We propose that the synergy of co-activator function between p300, CARM1 and PRMT1 is due to their different but complementary protein modification activities.


Subject(s)
Cell Nucleus/metabolism , Histones/metabolism , Transcriptional Activation , Acetylation , Animals , Arginine/metabolism , Cell Line , Chromatin/metabolism , Enhancer Elements, Genetic , Genes, Reporter , Intracellular Signaling Peptides and Proteins , Methylation , Methyltransferases/metabolism , Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Recombinant Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Transfection
16.
Mol Biol Cell ; 11(6): 2007-18, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848625

ABSTRACT

Hic-5 (hydrogen peroxide-inducible clone-5) is a focal adhesion protein that is involved in cellular senescence. In the present study, a yeast two-hybrid screen identified Hic-5 as a protein that interacts with a region of the glucocorticoid receptor that includes a nuclear matrix-targeting signal and the tau2 transcriptional activation domain. In transiently transfected mammalian cells, overexpression of Hic-5 potentiated the activation of reporter genes by all steroid receptors, excluding the estrogen receptor. The activity of the estrogen receptor and the thyroid hormone receptor was stimulated by Hic-5 in the presence but not in the absence of coexpressed coactivator GRIP1. In biochemical fractionations and indirect immunofluorescence assays, a fraction of endogenous Hic-5 in REF-52 cells and transiently expressed Hic-5 in Cos-1 cells was associated with the nuclear matrix. The C-terminal region of Hic-5, which contains seven zinc fingers arranged in four LIM domains, was required for interaction with focal adhesions, the nuclear matrix, steroid receptors, and the tau2 domain of glucocorticoid receptor. The N-terminal region of Hic-5 possesses a transcriptional activation domain and was essential for the coactivator activity of Hic-5. Given the coexisting cytoplasmic and nuclear distributions of Hic-5 and its role in steroid receptor-mediated transcriptional activation, it is proposed that Hic-5 might transmit signals that emanate at cell attachment sites and regulate transcription factors, such as steroid receptors.


Subject(s)
Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Zinc Fingers , Animals , Binding Sites , COS Cells , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Mice , Nuclear Matrix/metabolism , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, Progesterone/metabolism
17.
Mol Cell Biol ; 20(5): 1855-67, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10669760

ABSTRACT

Transcriptional activation by nuclear hormone receptors is mediated by the 160-kDa family of nuclear receptor coactivators. These coactivators associate with DNA-bound nuclear receptors and transmit activating signals to the transcription machinery through two activation domains. In screening for mammalian proteins that bind the C-terminal activation domain of the nuclear receptor coactivator GRIP1, we identified a new variant of mouse Zac1 which we call mZac1b. Zac1 was previously discovered as a putative transcriptional activator involved in regulation of apoptosis and the cell cycle. In yeast two-hybrid assays and in vitro, mZac1b bound to GRIP1, to CREB-binding protein (CBP) and p300 (which are coactivators for nuclear receptors and other transcriptional activators), and to nuclear receptors themselves in a hormone-independent manner. In transient-transfection assays mZac1b exhibited a transcriptional activation activity when fused with the Gal4 DNA binding domain, and it enhanced transcriptional activation by the Gal4 DNA binding domain fused to GRIP1 or CBP fragments. More importantly, mZac1b was a powerful coactivator for the hormone-dependent activity of nuclear receptors, including androgen, estrogen, glucocorticoid, and thyroid hormone receptors. However, with some reporter genes and in some cell lines mZac1b acted as a repressor rather than a coactivator of nuclear receptor activity. Thus, mZac1b can interact with nuclear receptors and their coactivators and play both positive and negative roles in regulating nuclear receptor function.


Subject(s)
Cell Cycle Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Trans-Activators/genetics , Transcription Factors , Transcription, Genetic , Animals , Gene Expression Regulation , Genes, Tumor Suppressor , HeLa Cells , Humans , Mice , Tumor Suppressor Proteins
18.
Hum Mol Genet ; 9(2): 267-74, 2000 Jan 22.
Article in English | MEDLINE | ID: mdl-10607837

ABSTRACT

Normal polymorphic size variation of the exon 1 CAG microsatellite of the androgen receptor (AR) is associated with prostate cancer, benign prostatic hyperplasia and male infertility. Furthermore, abnormal expansion of the satellite leads to Kennedy's disease. We have shown recently that the AR N-terminal domain (NTD), which contains the polyglutamine (polyQ) stretch (encoded by the CAG repeat), functionally interacts with the C-termini of p160 coactivators. In the present study we explored possible AR CAG size effects on the p160 coactivator-mediated transactivation activity of the receptor. First, we mapped the p160 coactivator interaction on the AR NTD and found an interaction surface between amino acids 351 and 537. Although this region is 'downstream' from the polyQ stretch, it is still within the AR NTD, is implicated in constitutive transactivation activity of the receptor, and thus might be subject to polyQ size modulation. Indeed, cotrans- fection experiments in cultured prostate epithelial cells, using AR constructs of varying CAG sizes and p160 coactivator expression vectors, revealed that increased polyQ length, up to a size of 42 repeats, inhibited both basal and coactivator-mediated AR transactivation activity. AR expression in these cells, on the other hand, was unaffected by the same increased CAG repeat size range. We conclude that the AR NTD contributes to AR transactivation activity via functional interactions with p160 coactivators and that increasing polyQ length negatively affects p160-mediated coactivation of the AR. This molecular mechanism thus might explain, at least in part, the observed phenotypic effects of the AR CAG size polymorphism.


Subject(s)
Peptides/metabolism , Receptors, Androgen/metabolism , Transcription Factors/antagonists & inhibitors , Androgen Receptor Antagonists , Binding Sites/genetics , Histone Acetyltransferases , Humans , Male , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2 , Peptides/genetics , Peptides/physiology , Receptors, Androgen/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics , Tumor Cells, Cultured , tau Proteins/genetics
19.
Mol Endocrinol ; 13(12): 2151-62, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598588

ABSTRACT

In this report, we demonstrate that, in contrast to most previously characterized nuclear receptors, hERR1 and hERR2 (human estrogen receptor-related protein 1 and -2) are constitutive activators of the classic estrogen response element (ERE) as well as the palindromic thyroid hormone response element (TRE(pal)) but not the glucocorticoid response element (GRE). This intrinsically activated state of hERR1 and hERR2 resides in the ligand-binding domains of the two genes and is transferable to a heterologous receptor. In addition, we show that members of the p160 family of nuclear receptor coactivators, ACTR (activator of thyroid and retinoic acid receptors), GRIP1 (glucocorticoid receptor interacting protein 1), and SRC-1 (steroid receptor coactivator 1), potentiate the transcriptional activity by hERR1 and hERR2 in mammalian cells, and that both orphan receptors bind the coactivators in a ligand-independent manner. Together, these results suggest that hERR1 and hERR2 activate gene transcription through a mechanism different from most of the previously characterized steroid hormone receptors.


Subject(s)
Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Transcription Factors/pharmacology , Transcription, Genetic , Acetyltransferases/metabolism , Acetyltransferases/pharmacology , Cell Line , Drug Synergism , Estrogens/pharmacology , HeLa Cells , Histone Acetyltransferases , Humans , Nuclear Receptor Coactivator 1 , Nuclear Receptor Coactivator 2 , Receptors, Cytoplasmic and Nuclear/genetics , Response Elements , Thyroid Hormones/pharmacology , ERRalpha Estrogen-Related Receptor
20.
Mol Cell Biol ; 19(10): 6509-22, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490591

ABSTRACT

Transcription factors, such as nuclear receptors, often exist in various forms that are generated by highly conserved splicing events. Whereas the functional significance of these splicing variants is often not known, it is known that nuclear receptors activate transcription through interaction with coactivators. The parameters, other than ligands, that might modulate those interactions, however, are not well characterized, nor is the role of splicing variants. In this study, transient transfection, yeast two-hybrid, and GST pulldown assays are used to show not only that nuclear receptor hepatocyte nuclear factor 4 alpha1 (HNF4alpha1, NR2A1) interacts with GRIP1, and other coactivators, in the absence of ligand but also that the uncommonly large F domain in the C terminus of the receptor inhibits that interaction. In vitro, the F domain was found to obscure an AF-2-independent binding site for GRIP1 that did not map to nuclear receptor boxes II or III. The results also show that a natural splicing variant containing a 10-amino-acid insert in the middle of the F domain (HNF4alpha2) abrogates that inhibition in vivo and in vitro. A series of protease digestion assays indicates that there may be structural differences between HNF4alpha1 and HNF4alpha2 in the F domain as well as in the ligand binding domain (LBD). The data also suggest that there is a direct physical contact between the F domain and the LBD of HNF4alpha1 and -alpha2 and that that contact is different in the HNF4alpha1 and HNF4alpha2 isoforms. Finally, we propose a model in which the F domain of HNF4alpha1 acts as a negative regulatory region for transactivation and in which the alpha2 insert ameliorates the negative effect of the F domain. A conserved repressor sequence in the F domains of HNF4alpha1 and -alpha2 suggests that this model may be relevant to other nuclear receptors as well.


Subject(s)
Alternative Splicing , DNA-Binding Proteins , Phosphoproteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Endopeptidases/metabolism , Hepatocyte Nuclear Factor 4 , Intracellular Signaling Peptides and Proteins , Models, Genetic , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 2 , Nuclear Receptor Coactivators , Phosphoproteins/genetics , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...