Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Histol Histopathol ; 39(3): 319-331, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37377225

ABSTRACT

Since the approval of brentuximab vedotin (BV), assessment of CD30 status by immunohistochemistry gained increasing importance in the clinical management of patients diagnosed with CD30-expressing lymphomas, including classical Hodgkin lymphoma (CHL). Paradoxically, patients with low or no CD30 expression respond to BV. This discrepancy may be due to lack of standardization in CD30 staining methods. In this study, we examined 29 cases of CHL and 4 cases of nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) for CD30 expression using a staining protocol that was designed to detect low CD30 expression levels, and an evaluation system similar to the Allred scoring system used for breast cancer evaluation. For CHL, 10% of cases had low scores and 3% were CD30 negative, with 3 cases in which the majority of tumor cells showed very weak staining. Unexpectedly, one of four cases of NLPHL was positive. We demonstrate intra-patient heterogeneity in CD30 expression levels and staining patterns in tumor cells. Three CHL cases with weak staining may have been missed without the use of control tissue for low expression. Thus, standardization of CD30 immunohistochemical staining with use of known low-expressing controls may aid in proper CD30 assessment and subsequent therapeutic stratification of patients.


Subject(s)
Hodgkin Disease , Humans , Brentuximab Vedotin/therapeutic use , Diagnosis, Differential , Hodgkin Disease/diagnosis , Hodgkin Disease/drug therapy , Hodgkin Disease/pathology , Immunohistochemistry , Staining and Labeling
2.
Toxicol Appl Pharmacol ; 339: 110-120, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29229235

ABSTRACT

Valproic acid (VPA) is a well-established therapeutic used in treatment of seizure and mood disorders as well as migraines and a known hepatotoxicant. About 50% of VPA users experience metabolic disruptions, including weight gain, hyperlipidemia, and hyperinsulinemia, among others. Several of these metabolic abnormalities are similar to the effects of circadian rhythm disruption. In the current study, we examine the effect of VPA exposure on the expression of core circadian transcription factors that drive the circadian clock via a transcription-translation feedback loop. In cells with an unsynchronized clock, VPA simultaneously upregulated the expression of genes encoding core circadian transcription factors that regulate the positive and negative limbs of the feedback loop. Using low dose glucocorticoid, we synchronized cultured fibroblast cells to a circadian oscillatory pattern. Whether VPA was added at the time of synchronization or 12h later at CT12, we found that VPA disrupted the oscillatory expression of multiple genes encoding essential transcription factors that regulate circadian rhythm. Therefore, we conclude that VPA has a potent effect on the circadian rhythm transcription-translation feedback loop that may be linked to negative VPA side effects in humans. Furthermore, our study suggests potential chronopharmacology implications of VPA usage.


Subject(s)
Anticonvulsants/toxicity , Circadian Rhythm/drug effects , Transcription Factors/biosynthesis , Transcriptional Activation/drug effects , Valproic Acid/toxicity , Animals , Cell Line, Tumor , Circadian Rhythm/physiology , Dose-Response Relationship, Drug , Gene Expression , Mice , NIH 3T3 Cells , Transcription Factors/genetics , Transcriptional Activation/physiology
3.
J Steroid Biochem Mol Biol ; 167: 1-13, 2017 03.
Article in English | MEDLINE | ID: mdl-27645313

ABSTRACT

Small molecule inhibitors of lysine deacetylases (KDACs) are approved for clinical use in treatment of several diseases. Nuclear receptors, such as the glucocorticoid receptor (GR) use lysine acetyltransferases (KATs or HATs) and KDACs to regulate transcription through acetylation and deacetylation of protein targets such as histones. Previously we have shown that KDAC1 activity facilitates GR-activated transcription at about half of all cellular target genes. In the current study we examine the role of Class I KDACs in glucocorticoid-mediated repression of gene expression. Inhibition of KDACs through two structurally distinct Class I-selective inhibitors prevented dexamethasone (Dex)-mediated transcriptional repression in a gene-selective fashion. In addition, KDAC activity is also necessary to maintain repression. Steroid receptor coactivator 2 (SRC2), which is known to play a vital role in GR-mediated repression of pro-inflammatory genes, was found to be dispensable for repression of glucocorticoid target genes sensitive to KDAC inhibition. At the promoters of these genes, KDAC inhibition did not result in altered nucleosome occupancy or histone H3 acetylation. Surprisingly, KDAC inhibition rapidly induced a significant decrease in H3K4Me2 at promoter nucleosomes with no corresponding change in H3K4Me3, suggesting the activation of the lysine demethylase, LSD1/KDM1A. Depletion of LSD1 expression via siRNA restored Dex-mediated repression in the presence of KDAC inhibitors, suggesting that LSD1 activation at these gene promoters is incompatible with transcriptional repression. Treatment with KDAC inhibitors does not alter cellular levels of LSD1 or its association with Dex-repressed gene promoters. Therefore, we conclude that Class I KDACs facilitate Dex-induced transcriptional repression by suppressing LSD1 complex activity at selected target gene promoters. Rather than facilitating repression of transcription, LSD1 opposes it in these gene contexts.


Subject(s)
Gene Expression Regulation , Glucocorticoids/metabolism , Histone Deacetylases/metabolism , Histone Demethylases/metabolism , Lysine/metabolism , Animals , Cell Line, Tumor , Dexamethasone/chemistry , Histones/metabolism , Inflammation , Mice , Nuclear Receptor Coactivator 2/metabolism , Nucleosomes/metabolism , Promoter Regions, Genetic , RNA/analysis , RNA, Small Interfering/metabolism
4.
Biochem Pharmacol ; 122: 62-71, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27836672

ABSTRACT

The molecular mechanisms behind the transition from simple steatosis to nonalcoholic steatohepatitis (NASH) in nonalcoholic fatty liver disease (NAFLD) are not clearly understood. This hinders development of effective therapies for treatment and prevention of NASH. In this study expression profiling data from normal, steatosis, and NASH human livers were used to predict transcription factors that are misregulated as mechanistic features of NAFLD progression. Previously-published human NAFLD gene expression profiling data from normal, steatosis, and NASH livers were subjected to transcription factor binding site enrichment analysis. Selected transcription factors that bind enriched transcription factor binding sites were analyzed for changes in expression. Distinct transcription factor binding sites were enriched in genes significantly up- or down-regulated in NASH livers. Those enriched in up-regulated genes were bound by transcription factors such as FOXA, CEBP, and HNF1 family members, while those enriched in down-regulated genes were bound by nuclear receptors involved in xenobiotic sensing and lipid metabolism. Levels of mRNA and protein for selected transcription factors were significantly changed during disease progression. The study indicates that NAFLD progression involves changes in activity or expression of transcription factors that regulate genes involved in hepatic processes known to be altered in NASH. Transcription factors such as PPAR receptors, FoxA family members, and HNF4A might be targeted therapeutically to prevent NAFLD progression.


Subject(s)
Gene Expression Regulation/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Transcription Factors/metabolism , Binding Sites , Disease Progression , Down-Regulation , Gene Expression Profiling , Humans , Protein Binding , Up-Regulation
5.
Cancer Biol Ther ; 17(12): 1240-1252, 2016 12.
Article in English | MEDLINE | ID: mdl-27791595

ABSTRACT

Diffuse Large B-cell lymphoma (DLBCL) is an aggressive malignancy that has a 60 percent 5-year survival rate, highlighting a need for new therapeutic approaches. Histone deacetylase inhibitors (HDACi) are novel therapeutics being clinically-evaluated in combination with a variety of other drugs. However, rational selection of companion therapeutics for HDACi is difficult due to their poorly-understood, cell-type specific mechanisms of action. To address this, we developed a pre-clinical model system of sensitivity and resistance to the HDACi belinostat using DLBCL cell lines. In the current study, we demonstrate that cell lines sensitive to the cytotoxic effects of HDACi undergo early mitotic arrest prior to apoptosis. In contrast, HDACi-resistant cell lines complete mitosis after a short delay and arrest in G1. To force mitotic arrest in HDACi-resistant cell lines, we used low dose vincristine or paclitaxel in combination with belinostat and observed synergistic cytotoxicity. Belinostat curtails vincristine-induced mitotic arrest and triggers a strong apoptotic response associated with downregulated MCL-1 expression and upregulated BIM expression. Resistance to microtubule targeting agents (MTAs) has been associated with their propensity to induce polyploidy and thereby increase the probability of genomic instability that enables cancer progression. Co-treatment with belinostat effectively eliminated a vincristine-induced, actively cycling polyploid cell population. Our study demonstrates that vincristine sensitizes DLBCL cells to the cytotoxic effects of belinostat and that belinostat prevents polyploidy that could cause vincristine resistance. Our findings provide a rationale for using low dose MTAs in conjunction with HDACi as a potential therapeutic strategy for treatment of aggressive DLBCL.


Subject(s)
Cytotoxins/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Mitosis/drug effects , Sulfonamides/pharmacology , Tubulin Modulators/pharmacology , Vincristine/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Drug Synergism , Humans , Models, Biological , Paclitaxel/pharmacology , Polyploidy , Up-Regulation
6.
Mol Carcinog ; 54(11): 1513-20, 2015 11.
Article in English | MEDLINE | ID: mdl-25307283

ABSTRACT

Sulforaphane is a natural product found in broccoli, which is known to exert many different molecular effects in the cell, including inhibition of histone deacetylase (HDAC) enzymes. Here, we examine for the first time the potential for sulforaphane to inhibit HDACs in HaCaT keratinocytes and compare our results with those found using HCT116 colon cancer cells. Significant inhibition of HDAC activity in HCT116 nuclear extracts required prolonged exposure to sulforaphane in the presence of serum. Under the same conditions HaCaT nuclear extracts did not exhibit reduced HDAC activity with sulforaphane treatment. Both cell types displayed down-regulation of HDAC protein levels by sulforaphane treatment. Despite these reductions in HDAC family member protein levels, acetylation of marker proteins (acetylated Histone H3, H4, and tubulin) was decreased by sulforaphane treatment. Time-course analysis revealed that HDAC6, HDAC3, and acetylated histone H3 protein levels are significantly inhibited as early as 6 h into sulforaphane treatment. Transcript levels of HDAC6 are also suppressed after 48 h of treatment. These results suggest that HDAC activity noted in nuclear extracts is not always translated as expected to target protein acetylation patterns, despite dramatic inhibition of some HDAC protein levels. In addition, our data suggest that keratinocytes are at least partially resistant to the nuclear HDAC inhibitory effects of sulforaphane, which is exhibited in HCT116 and other cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Isothiocyanates/pharmacology , Acetylation/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , HCT116 Cells , Histone Deacetylase 6 , Histones/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Sulfoxides
7.
Mol Endocrinol ; 28(5): 607-21, 2014 May.
Article in English | MEDLINE | ID: mdl-24645680

ABSTRACT

Lysine deacetylases have been known to regulate nuclear receptor function for many years. In the unliganded state, nuclear receptors that form heterodimers with retinoid X receptors, such as the retinoic acid and thyroid hormone receptors, associate with deacetylases to repress target genes. In the case of steroid receptors, binding of an antagonist ligand was initially reported to induce association of deacetylases to prevent activation of target genes. Since then, deacetylases have been shown to have diverse functions in steroid receptor signaling, from regulating interactions with molecular chaperones to facilitating their ability to activate transcription. The purpose of this review is to summarize recent studies on the role of deacetylases in steroid receptor signaling, which show deacetylases to be highly versatile regulators of steroid receptor function.


Subject(s)
Histone Deacetylases/physiology , Receptors, Steroid/metabolism , Acetylation , Animals , Epigenesis, Genetic , HSP90 Heat-Shock Proteins/metabolism , Humans , Protein Processing, Post-Translational , Signal Transduction
8.
J Biol Chem ; 288(40): 28900-12, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23946490

ABSTRACT

Nuclear receptors use lysine acetyltransferases and lysine deacetylases (KDACs) in regulating transcription through histone acetylation. Lysine acetyltransferases interact with steroid receptors upon binding of an agonist and are recruited to target genes. KDACs have been shown to interact with steroid receptors upon binding to an antagonist. We have shown previously that KDAC inhibitors (KDACis) potently repress the mouse mammary tumor virus promoter through transcriptional mechanisms and impair the ability of the glucocorticoid receptor (GR) to activate it, suggesting that KDACs can play a positive role in GR transactivation. In the current study, we extended this analysis to the entire GR transcriptome and found that the KDACi valproic acid impairs the ability of agonist-bound GR to activate about 50% of its target genes. This inhibition is largely due to impaired transcription rather than defective GR processing and was also observed using a structurally distinct KDACi. Depletion of KDAC1 expression mimicked the effects of KDACi in over half of the genes found to be impaired in GR transactivation. Simultaneous depletion of KDACs 1 and 2 caused full or partial impairment of several more GR target genes. Altogether we found that Class I KDAC activity facilitates GR-mediated activation at a sizable fraction of GR-activated target genes and that KDAC1 alone or in coordination with KDAC2 is required for efficient GR transactivation at many of these target genes. Finally, our work demonstrates that KDACi exposure has a significant impact on GR signaling and thus has ramifications for the clinical use of these drugs.


Subject(s)
Amidohydrolases/metabolism , Glucocorticoids/pharmacology , Lysine/metabolism , Transcription, Genetic/drug effects , Acetylation/drug effects , Animals , Cell Line, Tumor , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Histones/metabolism , Hydroxamic Acids/pharmacology , Mice , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Protein Binding/drug effects , Receptors, Glucocorticoid/metabolism , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Valproic Acid/chemistry , Valproic Acid/pharmacology
9.
Cancer Biol Ther ; 14(10): 949-61, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23982416

ABSTRACT

Diffuse large B cell lymphoma (DLBCL) is an aggressive form of non-Hodgkin lymphoma. While the initial treatment strategy is highly effective, relapse occurs in 40% of cases. Histone deacetylase inhibitors (HDACi) are a promising class of anti-cancer drugs but their single agent efficacy against relapsed DLBCL has been variable, ranging from few complete/partial responses to some stable disease. However, most patients showed no response to HDACi monotherapy for unknown reasons. Here we show that sensitivity and resistance to the hydroxamate HDACi, PXD101, can be modeled in DLBCL cell lines. Sensitivity is characterized by G 2/M arrest and apoptosis and resistance by reversible G 1 growth arrest. These responses to PXD101 are independent of several negative prognostic indicators such as DLBCL subtype, BCL2 and MYC co-expression, and p53 mutation, suggesting that HDACi might be used effectively against highly aggressive DLBCL tumors if they are combined with other therapeutics that overcome HDACi resistance. Our investigation of mechanisms underlying HDACi resistance showed that cyclin-dependent kinase inhibitors (CKIs), p21 and p27, are upregulated by PXD101 in a sustained fashion in resistant cell lines concomitant with decreased activity of the cyclin E/cdk2 complex and decreased Rb phosphorylation. PXD101 treatment results in increased association of CKI with the cyclin E/cdk2 complex in resistant cell lines but not in a sensitive line, indicating that the CKIs play a key role in G 1 arrest. The results suggest several treatment strategies that might increase the efficacy of HDACi against aggressive DLBCL.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Sulfonamides/pharmacology , Apoptosis , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitory Concentration 50 , Lymphoma, Large B-Cell, Diffuse/metabolism , Phosphorylation , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Retinoblastoma Protein/metabolism , Up-Regulation/drug effects
10.
J Biomed Biotechnol ; 2011: 416905, 2011.
Article in English | MEDLINE | ID: mdl-21253530

ABSTRACT

Histone deacetylases (HDACs) have been shown to be required for basal or inducible transcription at a variety of genes by poorly understood mechanisms. We demonstrated previously that HDAC inhibition rapidly repressed transcription from the mouse mammary tumor virus (MMTV) promoter by a mechanism that does not require the binding of upstream transcription factors. In the current study, we find that HDACs work through the core promoter sequences of MMTV as well as those of several cellular genes to facilitate transcriptional initiation through deacetylation of nonhistone proteins.


Subject(s)
Histone Deacetylases/metabolism , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic , Animals , Cell Line, Tumor , HeLa Cells , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Mammary Tumor Virus, Mouse/drug effects , Mice , Models, Genetic , RNA Polymerase II/metabolism , TATA Box/genetics , Transcription, Genetic/drug effects
11.
Mol Endocrinol ; 23(1): 61-73, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19036901

ABSTRACT

The progesterone receptor (PR) plays a pivotal role in proper development and function of the mammary gland and has also been implicated in mammary tumorigenesis. PR is a ligand-activated transcription factor; however, relatively, little is known about its mechanisms of action at endogenous target promoters. The aim of our study was to identify a natural PR-responsive gene and investigate its transcriptional regulation in the mammary microenvironment. Our experiments revealed FKBP5 as a direct target of the PR, because it exhibited a rapid activation by progestin that was cycloheximide independent and correlated with recruitment of RNA polymerase II to the promoter. Site-directed mutagenesis and chromatin immunoprecipitation assays showed that progestin responsiveness is mediated through a composite element in the first intron, to which the PR binds concomitantly with GATA-2. Mutational analysis of the element revealed that the GATA-2 site is essential for progestin activation. Direct binding of PR to DNA contributes to the efficiency of activation but is not sufficient, suggesting that the receptor makes important protein-protein interactions as part of its mechanism of action at the FKBP5 promoter. Using chromatin immunoprecipitation assays we also determined that the intronic region is in communication with the promoter, probably via DNA looping. Time course analysis revealed a cyclical pattern of PR recruitment to the FKBP5 gene but a persistent recruitment to the mouse mammary tumor virus promoter, indicating that receptor cycling is a gene-specific phenomenon rather than a characteristic of the receptor itself. Our study offers new insight in the nature of PR-regulated transcription in mammary cancer cells.


Subject(s)
GATA2 Transcription Factor/metabolism , Introns , Receptors, Progesterone/metabolism , Tacrolimus Binding Proteins/genetics , Animals , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , HeLa Cells , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse/genetics , Mice , Models, Biological , Mutagenesis, Site-Directed , Promegestone/pharmacology , Recombinant Proteins/genetics
12.
Exp Cell Res ; 314(15): 2855-69, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18644368

ABSTRACT

cAMP signaling is known to have significant effects on cell growth, either inhibitory or stimulatory depending on the cell type. Study of cAMP-induced growth inhibition in mammalian somatic cells has focused mainly on the combined role of protein kinase A (PKA) and mitogen-activated protein (MAP) kinases in regulation of progression through the G1 phase of the cell cycle. Here we show that cAMP signaling regulates histone H3 phosphorylation in a cell cycle-dependent fashion, increasing it in quiescent cells but dramatically reducing it in cycling cells. The latter is due to a rapid and dramatic loss of mitotic histone H3 phosphorylation caused by a disruption in G2 progression, as evidenced by the inhibition of mitotic entry and decreased activity of the CyclinB/Cdk1 kinase. The inhibition of G2 progression induced through cAMP signaling is dependent on expression of the catalytic subunit of PKA and is highly sensitive to intracellular cAMP concentration. The mechanism by which G2 progression is inhibited is independent of both DNA damage and MAP kinase signaling. Our results suggest that cAMP signaling activates a G2 checkpoint by a unique mechanism and provide new insight into normal cellular regulation of G2 progression.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclic AMP/metabolism , G2 Phase/genetics , Genes, cdc/physiology , Histones/metabolism , Mitosis/genetics , Animals , CDC2 Protein Kinase/metabolism , Catalytic Domain/physiology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin B/metabolism , Histones/genetics , Mice , NIH 3T3 Cells , Phosphorylation , Signal Transduction/genetics
13.
Bioessays ; 30(1): 15-24, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18081007

ABSTRACT

Transcriptional repression and silencing have been strongly associated with hypoacetylation of histones. Accordingly, histone deacetylases, which remove acetyl groups from histones, have been shown to participate in mechanisms of transcriptional repression. Therefore, current models of the role of acetylation in transcriptional regulation focus on the acetylation status of histones and designate histone acetyltransferases, which add acetyl groups to histones, as transcriptional coactivators and histone deacetylases as corepressors. In recent years, an accumulation of studies have shown that these enzymes also target non-histone proteins and that histone deacetylases have clear roles as coactivators at a variety of genes, some of which are key regulators of cell growth and survival. This review summarizes the evidence for histone deacetylases as coactivators and provides models of coactivation mechanisms, some of which integrate roles of acetylated histones and non-histone proteins in transcription.


Subject(s)
Histone Deacetylases/physiology , Transcriptional Activation/physiology , Animals , Humans , Models, Biological , Promoter Regions, Genetic , Trans-Activators/physiology , Transcription, Genetic/physiology
14.
Exp Cell Res ; 314(1): 1-10, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17950276

ABSTRACT

The phosphorylation of histone H3 is known to play a role in regulation of transcription as well as preparation of chromosomes for mitosis. Various signaling cascades induce H3 phosphorylation, particularly at genes activated by these pathways. In this study, we show that signaling can also have the opposite effect. Activators of cAMP signaling induce a rapid and potent loss of H3 phosphorylation. This effect is not mediated through a cAMP metabolite since a membrane-permeable form of AMP had no effect on H3 phosphorylation and a phosphodiesterase-resistant cAMP analog efficiently reduced it. cAMP is also the likely regulator of H3 phosphorylation under physiological conditions since only supra-pharmacological doses of cGMP induce the loss of H3 phosphorylation. The loss of phosphorylation is specific for histone H3 since we do not observe drastic losses in total phosphorylation of other histones. In addition, other H3 modifications are unaffected with the exception of lysine 9 methylation, which is elevated. Analysis of cell growth and cell cycle shows that cAMP signaling inhibits cell growth and arrests cells at both G1 and G2/M. Similar effects of cAMP signaling on H3 phosphorylation are observed in a variety of mammary adenocarcinoma-derived cell lines. In syngeneic human breast-derived cell lines, one diploid and non-transformed, the other derived from a ductal carcinoma, the loss of H3 phosphorylation is significantly more sensitive to cAMP concentration in the transformed cell line.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Cyclic AMP/metabolism , Histones/metabolism , Mammary Neoplasms, Animal/metabolism , Signal Transduction/genetics , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Breast Neoplasms/physiopathology , Carcinoma/physiopathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genes, cdc/physiology , Humans , Mammary Neoplasms, Animal/physiopathology , Methylation , Mice , Phosphorylation , Regulatory Elements, Transcriptional/genetics , Signal Transduction/drug effects
15.
J Biol Chem ; 279(15): 15231-9, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-14744870

ABSTRACT

Our previous work has shown that the progesterone receptor (PR) can exist in two distinct functional states in mammary adenocarcinoma cells. The differences in function included the ability to activate a promoter in organized chromatin, sensitivity to ligand, and ligand-independent activation. To determine whether these functional differences were because of altered cellular processing, we carried out biochemical analyses of the functionally distinct PRs. Although the majority of PR is localized to the nucleus, biochemical partitioning resulted in a loosely bound (cytosolic) fraction, and a tightly bound (nuclear) fraction. In the absence of progestins, the functionally distinct PRs differed significantly in partitioning between the two fractions. To characterize these fractions further, we analyzed interactions of unliganded PR with chaperones by coimmunoprecipitation. We determined that PR in the cytosolic fraction associated with hsp90 and p23. In contrast, PR in the nuclear fraction consisted of complexes containing hsp90, p23, and FKBP51 as well as PR that was dimerized and highly phosphorylated. Hormone treatment significantly reduced the formation of all PR-chaperone complexes. The hsp90 inhibitor, geldanamycin, similarly blocked transcriptional activity of both functionally distinct receptors. However, the two forms of the PR differed in their ability to associate with the mouse mammary tumor virus promoter in organized chromatin. These findings provide new information about the composition and distribution of mature progesterone receptor complexes in mammary adenocarcinoma cells, and suggest that differences in receptor subcellular distribution have a significant impact on their function. These findings also reveal that transiently expressed steroid receptors may not always be processed like their endogenous counterparts.


Subject(s)
Chromatin/metabolism , Receptors, Progesterone/chemistry , Animals , Benzoquinones , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Chromatin/chemistry , Cytosol/metabolism , DNA/chemistry , DNA/metabolism , Dimerization , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique, Indirect , Genes, Viral , HSP90 Heat-Shock Proteins/metabolism , Hormones/metabolism , In Situ Hybridization, Fluorescence , Intramolecular Oxidoreductases , Lactams, Macrocyclic , Ligands , Luciferases/metabolism , Mammary Tumor Virus, Mouse/genetics , Mice , Microscopy, Fluorescence , Models, Biological , Molecular Chaperones/metabolism , Phosphoproteins/metabolism , Phosphorylation , Precipitin Tests , Promoter Regions, Genetic , Prostaglandin-E Synthases , Protein Binding , Protein Conformation , Quinones/pharmacology , Receptors, Progesterone/metabolism , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/metabolism , Transcription, Genetic , Transfection , beta-Galactosidase/metabolism
16.
Oncogene ; 22(31): 4807-18, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12894222

ABSTRACT

Increased histone acetylation has been associated with activated gene transcription and decreased acetylation with repression. However, there is a growing number of genes known, which are downregulated by histone deacetylase (HDAC) inhibitors through unknown mechanisms. This study examines the mechanism by which the mouse mammary tumor virus (MMTV) promoter is repressed by the HDAC inhibitor, trichostatin A (TSA). We find that this repression is transcriptional in nature and that it occurs in the presence and absence of glucocorticoids. TSA decreases MMTV transcription at a rapid rate, reaching maximum in 30-60 min. In contrast with previous reports, the repression does not correlate with an inhibition of glucocorticoid-induced nuclease hypersensitivity or NF1-binding at the MMTV promoter. Surprisingly, TSA does not induce sizable increases in histone acetylation at the MMTV promoter nor does it inhibit histone deacetylation, which accompanies deactivation of the glucocorticoid-activated MMTV promoter. Repression of MMTV transcription by TSA does not depend on the chromatin organization of the promoter because a transiently transfected MMTV promoter construct with a disorganized nucleoprotein structure was also repressed by TSA treatment. Mutational analysis of the MMTV promoter indicates that repression by TSA is mediated through the TATA box region. These results suggest a novel mechanism that involves acetylation of nonhistone proteins necessary for basal transcription.


Subject(s)
Chromatin/ultrastructure , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Histones/metabolism , Hydroxamic Acids/pharmacology , Mammary Tumor Virus, Mouse/genetics , Protein Processing, Post-Translational/drug effects , Transcription, Genetic/drug effects , Acetylation/drug effects , Adenocarcinoma/pathology , Animals , Cell Transformation, Viral , Chromatin/drug effects , Dexamethasone/pharmacology , Female , Genes, Reporter , Mammary Neoplasms, Experimental/pathology , Mice , Nucleosomes/drug effects , Nucleosomes/ultrastructure , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/biosynthesis , Sequence Deletion , TATA Box , Terminal Repeat Sequences , Transfection
17.
Exp Cell Res ; 287(2): 361-73, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12837291

ABSTRACT

The nucleoprotein structure of the mouse mammary tumor virus (MMTV) promoter defines its response to cAMP signaling. A stably replicating MMTV template in highly organized chromatin is repressed in the presence of cAMP, whereas a transiently transfected template with a disorganized structure is activated. In this study, we investigate the nature of the cAMP-induced signal(s) by which these opposing responses occur to gain insight into their mechanism. We demonstrate that the transcriptional changes observed at both templates are mediated through cAMP-dependent protein kinase A (PKA). In addition, the MMTV promoter lacks a consensus cAMP response element (CRE) and neither template requires cAMP response element-binding protein (CREB) to elicit a response to cAMP signaling. However, the responses of the two templates differ mechanistically in that the CREB-binding protein p300 potentiates activation from the transient template in a manner dependent on its Cys/His-rich region 3, but does not appear to affect the repression of the replicating chromatin template. Chromatin immunoprecipitation assays show that cAMP treatment results in a decrease in acetylation of histone H4, and in multiple modifications of histone H3 at specific nucleosomes in the promoter region of the stable MMTV template. These findings suggest novel CREB-independent, chromatin-dependent pathways for transcriptional regulation by cAMP.


Subject(s)
Chromatin/metabolism , Cyclic AMP/metabolism , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic , Signal Transduction , Sulfonamides , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Acetylation , Animals , Cell Line , Chromatin/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Histones/metabolism , Isoquinolines/pharmacology , Kinetics , Mice , Nucleosomes/metabolism , RNA, Messenger/analysis , Receptors, Glucocorticoid/metabolism , Templates, Genetic , Transcriptional Activation
18.
J Biol Chem ; 277(31): 28247-55, 2002 Aug 02.
Article in English | MEDLINE | ID: mdl-12029095

ABSTRACT

The glucocorticoid receptor (GR) contains several activation domains, tau1 (AF-1), tau2, and AF-2, which were initially defined using transiently transfected reporter constructs. Using domain mutations in the context of full-length GR, this study defines those domains required for activation of the mouse mammary tumor virus (MMTV) promoter in two distinct nucleoprotein configurations. A transiently transfected MMTV template with a disorganized, accessible chromatin structure was largely dependent on the AF-2 domain for activation. In contrast, activation of an MMTV template in organized, replicated chromatin requires both domains but has a relatively larger dependence on the tau1 domain. Domain requirements for GR-induced chromatin remodeling of the latter template were also investigated. Mutation of the AF-2 helix 12 domain partially inhibits the induction of nuclease hypersensitivity, but the inhibition was relieved in the absence of tau1, suggesting the occurrence of an important interaction between the two domains. Further mutational analysis indicates that GR-induced chromatin remodeling requires the ligand-binding domain in the region of helix 3. Our study shows that the GR activation surfaces required for transcriptional modulation of a target promoter were determined in part by its chromatin structure. Within a particular cellular environment the GR appears to possess a significant degree of versatility in the mechanism by which it activates a target promoter.


Subject(s)
Chromatin/physiology , Mammary Tumor Virus, Mouse/genetics , Nucleoproteins/metabolism , Promoter Regions, Genetic , Receptors, Glucocorticoid/genetics , Alanine , Amino Acid Substitution , Animals , Base Sequence , Binding Sites , Chromatin/ultrastructure , Cloning, Molecular , Dexamethasone/pharmacology , Mice , Models, Molecular , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides , Protein Structure, Secondary , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Receptors, Interferon/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serine , Templates, Genetic , Transfection , Tumor Cells, Cultured
19.
J Biol Chem ; 277(22): 19847-54, 2002 May 31.
Article in English | MEDLINE | ID: mdl-11909860

ABSTRACT

Proteins encoded by the adenovirus E1A gene regulate both cellular and viral genes to mediate effects on cell cycle, differentiation, and cell growth control. We have identified the mouse mammary tumor virus (MMTV) promoter as a target of E1A action and investigated the role nucleoprotein structure plays in its response to E1A. Both 12 and 13 S forms target the MMTV promoter when it has a disorganized and accessible chromatin configuration. However, whereas the 13 S form is stimulatory, the 12 S form is repressive. When the MMTV promoter adopts an organized and repressed chromatin structure, it is targeted only by the 13 S form, which stimulates it. Although evidence indicates that E1A interacts with the SWI/SNF remodeling complex, E1A had no effect on chromatin remodeling at the MMTV promoter in organized chromatin. Analysis of E1A mutants showed that stimulation of the MMTV promoter is mediated solely through conserved region 3 and does not require interaction with Rb, p300/CBP-associated factor, or CBP/p300. Imaging analysis showed that E1A colocalizes with MMTV sequences in vivo, suggesting that it functions directly at the promoter. These results indicate that E1A stimulates the MMTV promoter in a fashion independent of chromatin conformation and through a direct mechanism involving interaction with the basal transcription machinery.


Subject(s)
Adenovirus E1A Proteins/metabolism , Adenovirus E1A Proteins/physiology , Mammary Tumor Virus, Mouse/genetics , Promoter Regions, Genetic , Animals , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , Fluorescent Antibody Technique, Indirect , Genetic Vectors , Luciferases/metabolism , Mice , Microscopy, Fluorescence , Mutation , Plasmids/metabolism , Protein Binding , Protein Isoforms , RNA/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...