Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3415, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649367

ABSTRACT

An important epigenetic component of tyrosine kinase signaling is the phosphorylation of histones, and epigenetic readers, writers, and erasers. Phosphorylation of protein arginine methyltransferases (PRMTs), have been shown to enhance and impair their enzymatic activity. In this study, we show that the hyperactivation of Janus kinase 2 (JAK2) by the V617F mutation phosphorylates tyrosine residues (Y149 and Y334) in coactivator-associated arginine methyltransferase 1 (CARM1), an important target in hematologic malignancies, increasing its methyltransferase activity and altering its target specificity. While non-phosphorylatable CARM1 methylates some established substrates (e.g. BAF155 and PABP1), only phospho-CARM1 methylates the RUNX1 transcription factor, on R223 and R319. Furthermore, cells expressing non-phosphorylatable CARM1 have impaired cell-cycle progression and increased apoptosis, compared to cells expressing phosphorylatable, wild-type CARM1, with reduced expression of genes associated with G2/M cell cycle progression and anti-apoptosis. The presence of the JAK2-V617F mutant kinase renders acute myeloid leukemia (AML) cells less sensitive to CARM1 inhibition, and we show that the dual targeting of JAK2 and CARM1 is more effective than monotherapy in AML cells expressing phospho-CARM1. Thus, the phosphorylation of CARM1 by hyperactivated JAK2 regulates its methyltransferase activity, helps select its substrates, and is required for the maximal proliferation of malignant myeloid cells.


Subject(s)
Apoptosis , Core Binding Factor Alpha 2 Subunit , Janus Kinase 2 , Protein-Arginine N-Methyltransferases , Tyrosine , Humans , Phosphorylation , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Protein-Arginine N-Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Tyrosine/metabolism , Cell Line, Tumor , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Methylation , Substrate Specificity , HEK293 Cells , Cell Cycle , Mutation
2.
bioRxiv ; 2023 Aug 11.
Article in English | MEDLINE | ID: mdl-37609261

ABSTRACT

Cell-intrinsic mechanisms of immunogenicity in ovarian cancer (OC) are not well understood. The presence of damaging mutations in the SWI/SNF chromatin remodeling complex, such as the SMARCA4 (BRG1) catalytic subunit, has been associated with improved response to ICB, however the mechanism by which this occurs is unclear. The aim of this current study was to examine the alterations in tumor cell-intrinsic and extrinsic immune signaling caused by SMARCA4 loss. Using OC models with loss-of-function mutations in SMARCA4 , we found that SMARCA4 loss resulted in increased cancer cell-intrinsic immunogenicity, characterized by upregulation of long-terminal RNA repeats such as endogenous retroviruses, increased expression of interferon-stimulated genes, and upregulation of antigen presentation machinery. Notably, this response was dependent on IRF3 signaling, but was independent of the type I interferon receptor. Mice inoculated with cancer cells bearing SMARCA4 loss demonstrated increased activation of cytotoxic T cells and NK cells in the tumor microenvironment as well as increased infiltration with activated dendritic cells. These results were recapitulated when animals bearing SMARCA4- proficient tumors were treated with a BRG1 inhibitor, suggesting that modulation of chromatin remodeling through targeting SMARCA4 may serve as a strategy to reverse immune evasion in OC.

3.
JCI Insight ; 6(19)2021 10 08.
Article in English | MEDLINE | ID: mdl-34622806

ABSTRACT

Myelodysplastic syndromes (MDS) are hematopoietic stem and progenitor cell (HSPC) malignancies characterized by ineffective hematopoiesis and an increased risk of leukemia transformation. Epigenetic regulators are recurrently mutated in MDS, directly implicating epigenetic dysregulation in MDS pathogenesis. Here, we identified a tumor suppressor role of the acetyltransferase p300 in clinically relevant MDS models driven by mutations in the epigenetic regulators TET2, ASXL1, and SRSF2. The loss of p300 enhanced the proliferation and self-renewal capacity of Tet2-deficient HSPCs, resulting in an increased HSPC pool and leukemogenicity in primary and transplantation mouse models. Mechanistically, the loss of p300 in Tet2-deficient HSPCs altered enhancer accessibility and the expression of genes associated with differentiation, proliferation, and leukemia development. Particularly, p300 loss led to an increased expression of Myb, and the depletion of Myb attenuated the proliferation of HSPCs and improved the survival of leukemia-bearing mice. Additionally, we show that chemical inhibition of p300 acetyltransferase activity phenocopied Ep300 deletion in Tet2-deficient HSPCs, whereas activation of p300 activity with a small molecule impaired the self-renewal and leukemogenicity of Tet2-deficient cells. This suggests a potential therapeutic application of p300 activators in the treatment of MDS with TET2 inactivating mutations.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , p300-CBP Transcription Factors/genetics , Animals , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Disease Models, Animal , Disease Progression , Epigenesis, Genetic , Hematopoietic Stem Cells , Leukemia, Myeloid, Acute/metabolism , Mice , Mutation , Myelodysplastic Syndromes/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Repressor Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Survival Rate
4.
Nucleic Acids Res ; 48(6): 2956-2968, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32025719

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) catalyzes the symmetric di-methylation of arginine residues in histones H3 and H4, marks that are generally associated with transcriptional repression. However, we found that PRMT5 inhibition or depletion led to more genes being downregulated than upregulated, indicating that PRMT5 can also act as a transcriptional activator. Indeed, the global level of histone H3K27me3 increases in PRMT5 deficient cells. Although PRMT5 does not directly affect PRC2 enzymatic activity, methylation of histone H3 by PRMT5 abrogates its subsequent methylation by PRC2. Treating AML cells with an EZH2 inhibitor partially restored the expression of approximately 50% of the genes that are initially downregulated by PRMT5 inhibition, suggesting that the increased H3K27me3 could directly or indirectly contribute to the transcription repression of these genes. Indeed, ChIP-sequencing analysis confirmed an increase in the H3K27me3 level at the promoter region of a quarter of these genes in PRMT5-inhibited cells. Interestingly, the anti-proliferative effect of PRMT5 inhibition was also partially rescued by treatment with an EZH2 inhibitor in several leukemia cell lines. Thus, PRMT5-mediated crosstalk between histone marks contributes to its functional effects.


Subject(s)
Arginine/metabolism , Histones/metabolism , Polycomb-Group Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Transcription, Genetic , Animals , Cell Cycle/genetics , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Methylation , Mice, Knockout , Models, Biological , Nucleosomes/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors
5.
Sci Rep ; 9(1): 6304, 2019 04 19.
Article in English | MEDLINE | ID: mdl-31004086

ABSTRACT

Here we demonstrate a mode of reciprocal regulation between GFI1 and p53 that controls the induction of apoptosis in T cells. We show that GFI1 prevents induction of p53 dependent apoptosis by recruiting LSD1 to p53, which leads to the demethylation of its C-terminal domain. This is accompanied by a decrease of the acetylation of lysine 117 within the core domain of the murine p53 protein, which is required for transcriptional induction of apoptosis. Our results support a model in which the effect of GFI1's regulation of methylation at the c-terminus of p53 is ultimately mediated through control of acetylation at lysine 117 of p53. We propose that GFI1 acts prior to the occurrence of DNA damage to affect the post-translational modification state and limit the subsequent activation of p53. Once activated, p53 then transcriptionally activates GFI1, presumably in order to re-establish the homeostatic balance of p53 activity. These findings have implications for the activity level of p53 in various disease contexts where levels of GFI1 are either increased or decreased.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , T-Lymphocytes/cytology , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
8.
Cell Rep ; 24(10): 2643-2657, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30184499

ABSTRACT

Protein arginine methyltransferase 5 (PRMT5) is overexpressed in many cancer types and is a promising therapeutic target for several of them, including leukemia and lymphoma. However, we and others have reported that PRMT5 is essential for normal physiology. This dependence may become dose limiting in a therapeutic setting, warranting the search for combinatorial approaches. Here, we report that PRMT5 depletion or inhibition impairs homologous recombination (HR) DNA repair, leading to DNA-damage accumulation, p53 activation, cell-cycle arrest, and cell death. PRMT5 symmetrically dimethylates histone and non-histone substrates, including several components of the RNA splicing machinery. We find that PRMT5 depletion or inhibition induces aberrant splicing of the multifunctional histone-modifying and DNA-repair factor TIP60/KAT5, which selectively affects its lysine acetyltransferase activity and leads to impaired HR. As HR deficiency sensitizes cells to PARP inhibitors, we demonstrate here that PRMT5 and PARP inhibitors have synergistic effects on acute myeloid leukemia cells.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Alternative Splicing/genetics , Alternative Splicing/physiology , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Death , Cell Line, Tumor , DNA Repair/genetics , DNA Repair/physiology , Histone Code/genetics , Histone Code/physiology , Histones/metabolism , Humans , Lysine Acetyltransferase 5/genetics , Lysine Acetyltransferase 5/metabolism , Lysine Acetyltransferases/genetics , Lysine Acetyltransferases/metabolism , Protein-Arginine N-Methyltransferases/genetics
9.
Cancer Cell ; 33(6): 1111-1127.e5, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894694

ABSTRACT

Chromatin-modifying enzymes, and specifically the protein arginine methyltransferases (PRMTs), have emerged as important targets in cancer. Here, we investigated the role of CARM1 in normal and malignant hematopoiesis. Using conditional knockout mice, we show that loss of CARM1 has little effect on normal hematopoiesis. Strikingly, knockout of Carm1 abrogates both the initiation and maintenance of acute myeloid leukemia (AML) driven by oncogenic transcription factors. We show that CARM1 knockdown impairs cell-cycle progression, promotes myeloid differentiation, and ultimately induces apoptosis. Finally, we utilize a selective, small-molecule inhibitor of CARM1 to validate the efficacy of CARM1 inhibition in leukemia cells in vitro and in vivo. Collectively, this work suggests that targeting CARM1 may be an effective therapeutic strategy for AML.


Subject(s)
Gene Expression Regulation, Leukemic , Hematopoiesis/genetics , Leukemia, Myeloid/genetics , Protein-Arginine N-Methyltransferases/genetics , Acute Disease , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Protein-Arginine N-Methyltransferases/metabolism
11.
J Clin Invest ; 125(9): 3532-44, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258414

ABSTRACT

Epigenetic regulators play critical roles in normal hematopoiesis, and the activity of these enzymes is frequently altered in hematopoietic cancers. The major type II protein arginine methyltransferase PRMT5 catalyzes the formation of symmetric dimethyl arginine and has been implicated in various cellular processes, including pluripotency and tumorigenesis. Here, we generated Prmt5 conditional KO mice to evaluate the contribution of PRMT5 to adult hematopoiesis. Loss of PRMT5 triggered an initial but transient expansion of hematopoietic stem cells (HSCs); however, Prmt5 deletion resulted in a concurrent loss of hematopoietic progenitor cells (HPCs), leading to fatal BM aplasia. PRMT5-specific effects on hematopoiesis were cell intrinsic and depended on PRMT5 methyltransferase activity. We found that PRMT5-deficient hematopoietic stem and progenitor cells exhibited severely impaired cytokine signaling as well as upregulation of p53 and expression of its downstream targets. Together, our results demonstrate that PRMT5 plays distinct roles in the behavior of HSCs compared with HPCs and is essential for the maintenance of adult hematopoietic cells.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/enzymology , Protein Methyltransferases/metabolism , Signal Transduction/physiology , Animals , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Protein Methyltransferases/genetics , Protein-Arginine N-Methyltransferases , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/physiology
12.
Genes Dev ; 27(17): 1868-85, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24013501

ABSTRACT

The p53 tumor suppressor is a transcription factor that mediates varied cellular responses. The C terminus of p53 is subjected to multiple and diverse post-translational modifications. An attractive hypothesis is that differing sets of combinatorial modifications therein determine distinct cellular outcomes. To address this in vivo, a Trp53(ΔCTD/ΔCTD) mouse was generated in which the endogenous p53 is targeted and replaced with a truncated mutant lacking the C-terminal 24 amino acids. These Trp53(ΔCTD/ΔCTD) mice die within 2 wk post-partum with hematopoietic failure and impaired cerebellar development. Intriguingly, the C terminus acts via three distinct mechanisms to control p53-dependent gene expression depending on the tissue. First, in the bone marrow and thymus, the C terminus dampens p53 activity. Increased senescence in the Trp53(ΔCTD/ΔCTD) bone marrow is accompanied by up-regulation of Cdkn1 (p21). In the thymus, the C-terminal domain negatively regulates p53-dependent gene expression by inhibiting promoter occupancy. Here, the hyperactive p53(ΔCTD) induces apoptosis via enhanced expression of the proapoptotic Bbc3 (Puma) and Pmaip1 (Noxa). In the liver, a second mechanism prevails, since p53(ΔCTD) has wild-type DNA binding but impaired gene expression. Thus, the C terminus of p53 is needed in liver cells at a step subsequent to DNA binding. Finally, in the spleen, the C terminus controls p53 protein levels, with the overexpressed p53(ΔCTD) showing hyperactivity for gene expression. Thus, the C terminus of p53 regulates gene expression via multiple mechanisms depending on the tissue and target, and this leads to specific phenotypic effects in vivo.


Subject(s)
Gene Expression Regulation , Genes, p53/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cellular Senescence/genetics , Cerebellum/growth & development , Cerebellum/metabolism , Gene Knock-In Techniques , Growth and Development/genetics , Liver/metabolism , Mice , Mice, Inbred C57BL , Mutation/genetics , Protein Binding , Protein Processing, Post-Translational , Sequence Deletion/genetics , Thymocytes/cytology , Thymocytes/metabolism , Time Factors
13.
Genes Dev ; 26(14): 1533-45, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22802528

ABSTRACT

The p53 tumor suppressor protein is a transcription factor that exerts its effects on the cell cycle via regulation of gene expression. Although the mechanism of p53-dependent transcriptional activation has been well-studied, the molecular basis for p53-mediated repression has been elusive. The E2F family of transcription factors has been implicated in regulation of cell cycle-related genes, with E2F6, E2F7, and E2F8 playing key roles in repression. In response to cellular DNA damage, E2F7, but not E2F6 or E2F8, is up-regulated in a p53-dependent manner, with p53 being sufficient to increase expression of E2F7. Indeed, p53 occupies the promoter of the E2F7 gene after genotoxic stress, consistent with E2F7 being a novel p53 target. Ablation of E2F7 expression abrogates p53-dependent repression of a subset of its targets, including E2F1 and DHFR, in response to DNA damage. Furthermore, E2F7 occupancy of the E2F1 and DHFR promoters is detected, and expression of E2F7 is sufficient to inhibit cell proliferation. Taken together, these results show that p53-dependent transcriptional up-regulation of its target, E2F7, leads to repression of relevant gene expression. In turn, this E2F7-dependent mechanism contributes to p53-dependent cell cycle arrest in response to DNA damage.


Subject(s)
DNA Damage , E2F7 Transcription Factor/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , E2F7 Transcription Factor/genetics , Humans , Response Elements , Tetrahydrofolate Dehydrogenase/biosynthesis , Tetrahydrofolate Dehydrogenase/genetics , Tumor Suppressor Protein p53/genetics
14.
J Biol Chem ; 287(26): 22397-407, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22514277

ABSTRACT

The p53 gene encodes a transcription factor that is composed of several functional domains: the N-terminal transactivation domain, the central sequence-specific DNA binding domain, the tetramerization domain, and the highly basic C-terminal regulatory domain (CTD). The p53 CTD is a nonspecific DNA binding domain that is subject to extensive post-translational modifications. However, the functional significance of the p53 CTD remains unclear. The role of this domain in the regulation of p53 functions is explored by comparing the activity of ectopically expressed wild-type (WT) p53 protein to that of a truncated mutant lacking the 24 terminal amino acids (Δ24). Using quantitative real time PCR and chromatin Immuno-Precipitation experiments, a p53 CTD deletion is shown to alter the p53-dependent induction of a subset of its target genes due to impaired specific DNA binding. Moreover, p53-induced growth arrest and apoptosis both require an intact p53 CTD. These data indicate that the p53 CTD is a positive regulator of p53 tumor suppressor functions.


Subject(s)
Gene Expression Regulation , Tumor Suppressor Protein p53/chemistry , Amino Acids/chemistry , Apoptosis , Bromodeoxyuridine/pharmacology , Cell Line , Cell Line, Tumor , Cell Lineage , Chromatin Immunoprecipitation , DNA/metabolism , Humans , Microscopy, Fluorescence/methods , Plasmids/metabolism , Protein Binding , Protein Structure, Tertiary , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
15.
Cancer Cell ; 21(1): 3-5, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22264782

ABSTRACT

In this issue of Cancer Cell, Gajjar et al. provide insight into how Mdm2 can both inhibit and enhance p53 activity. In the basal setting, Mdm2 binds p53 and promotes p53 degradation. Under stress conditions, ATM-dependent phosphorylation of Mdm2 results in its recruitment to p53 mRNA, thereby stimulating p53 translation.

16.
J Mol Biol ; 384(4): 980-91, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-18950637

ABSTRACT

The ubiquitous activating transcription factor (ATF) 7 binds as a homodimer to the cAMP response element/TPA response element motifs present in the promoters of its target genes. ATF7 is homologous to ATF2 and heterodimerizes with Jun or Fos proteins, modulating their DNA-binding specificities. We previously demonstrated that TAF12, a component of the TFIID general transcription factor, mediates ATF7 transcriptional activity through direct interactions between the two proteins. By contrast, ATF7, but not ATF2, is modified in vivo by sumoylation, which restricts its subcellular localization, thereby inhibiting its transcriptional activity. In the present study, we dissect the mechanism of this functional switch. We characterized the multisite phosphorylation of the ATF7 activation domain and identified one of the involved kinase, p38beta2 mitogen-activated protein kinase. In addition, we show that epidermal growth factor treatment results in a two-step modification mechanism of ATF7 activation domain. The Thr53 residue is phosphorylated first by a presently unknown kinase, allowing p38beta2 mitogen-activated protein kinase to modify the Thr51 residue, excluding the sumoylation of ATF7 protein. The resulting activation of transcription is related to an increased association of TAF12 with this phosphorylated form of ATF7. Our data therefore conclusively establish that sumoylation and phosphorylation of ATF7 are two antagonistic posttranslational modifications.


Subject(s)
Activating Transcription Factors/metabolism , Mitogen-Activated Protein Kinase 11/metabolism , Phosphorylation , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription, Genetic
17.
Nucleic Acids Res ; 35(4): 1134-44, 2007.
Article in English | MEDLINE | ID: mdl-17264123

ABSTRACT

Over the past few years, small ubiquitin-like modifier (SUMO) modification has emerged as an important regulator of diverse pathways and activities including protein localization and transcriptional regulation. We identified a consensus sumoylation motif (IKEE), located within the N-terminal activation domain of the ATF7 transcription factor and thus investigated the role of this modification. ATF7 is a ubiquitously expressed transcription factor, homologous to ATF2, that binds to CRE elements within specific promoters. This protein is able to heterodimerize with Jun or Fos proteins and its transcriptional activity is mediated by interaction with TAF12, a subunit of the general transcription factor TFIID. In the present article, we demonstrate that ATF7 is sumoylated in vitro (using RanBP2 as a E3-specific ligase) and in vivo. Moreover, we show that ATF7 sumoylation affects its intranuclear localization by delaying its entry into the nucleus. Furthermore, SUMO conjugation inhibits ATF7 transactivation activity by (i) impairing its association with TAF12 and (ii) blocking its binding-to-specific sequences within target promoters.


Subject(s)
Activating Transcription Factors/metabolism , Protein Processing, Post-Translational , SUMO-1 Protein/metabolism , Activating Transcription Factors/analysis , Activating Transcription Factors/antagonists & inhibitors , Cell Line , Cell Nucleus/chemistry , Humans , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
Oncogene ; 24(21): 3472-83, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15735663

ABSTRACT

The ATF7 proteins, which are members of the cyclic AMP responsive binding protein (CREB)/activating transcription factor (ATF) family of transcription factors, display quite versatile properties: they can interact with the adenovirus E1a oncoprotein, mediating part of its transcriptional activity; they heterodimerize with the Jun, Fos or related transcription factors, likely modulating their DNA-binding specificity; they also recruit to the promoter a stress-induced protein kinase (JNK2). In the present study, we investigate the functional relationships of ATF7 with hsTAF12 (formerly hsTAF(II)20/15), which has originally been identified as a component of the general transcription factor TFIID. We show that overexpression of hsTAF12 potentiates ATF7-induced transcriptional activation through direct interaction with ATF7, suggesting that TAF12 is a functional partner of ATF7. In support of this conclusion, chromatin immunoprecipitation experiments confirm the interaction of ATF7 with TAF12 on an ATF7-responsive promoter, in the absence of any artificial overexpression of both proteins. We also show that the TAF12-dependent transcriptional activation is competitively inhibited by TAF4. Although both TAF12 isoforms (TAF12-1 and -2, formerly TAF(II)20 and TAF(II)15) interact with the ATF7 activation region through their histone-fold domain, only the largest, hsTAF12-1, mediates transcriptional activation through its N-terminal region.


Subject(s)
DNA-Binding Proteins/pharmacology , Gene Expression Regulation , TATA-Binding Protein Associated Factors/biosynthesis , TATA-Binding Protein Associated Factors/pharmacology , Transcription Factor TFIID/pharmacology , Transcription Factors/pharmacology , Activating Transcription Factors , Amino Acid Sequence , Animals , Cell Line , Chromatin , DNA-Binding Proteins/biosynthesis , Humans , Immunoprecipitation , Insecta , Leucine Zippers , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...