Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters











Publication year range
1.
Cell Death Dis ; 5: e1257, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24874729

ABSTRACT

The platinum derivative cis-diamminedichloroplatinum(II), best known as cisplatin, is currently employed for the clinical management of patients affected by testicular, ovarian, head and neck, colorectal, bladder and lung cancers. For a long time, the antineoplastic effects of cisplatin have been fully ascribed to its ability to generate unrepairable DNA lesions, hence inducing either a permanent proliferative arrest known as cellular senescence or the mitochondrial pathway of apoptosis. Accumulating evidence now suggests that the cytostatic and cytotoxic activity of cisplatin involves both a nuclear and a cytoplasmic component. Despite the unresolved issues regarding its mechanism of action, the administration of cisplatin is generally associated with high rates of clinical responses. However, in the vast majority of cases, malignant cells exposed to cisplatin activate a multipronged adaptive response that renders them less susceptible to the antiproliferative and cytotoxic effects of the drug, and eventually resume proliferation. Thus, a large fraction of cisplatin-treated patients is destined to experience therapeutic failure and tumor recurrence. Throughout the last four decades great efforts have been devoted to the characterization of the molecular mechanisms whereby neoplastic cells progressively lose their sensitivity to cisplatin. The advent of high-content and high-throughput screening technologies has accelerated the discovery of cell-intrinsic and cell-extrinsic pathways that may be targeted to prevent or reverse cisplatin resistance in cancer patients. Still, the multifactorial and redundant nature of this phenomenon poses a significant barrier against the identification of effective chemosensitization strategies. Here, we discuss recent systems biology studies aimed at deconvoluting the complex circuitries that underpin cisplatin resistance, and how their findings might drive the development of rational approaches to tackle this clinically relevant problem.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Systems Biology , Animals , Humans , Systems Biology/methods , Systems Biology/trends
2.
Br J Cancer ; 108(11): 2304-11, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23695020

ABSTRACT

BACKGROUND: Eukaryotic translation elongation factor 1A2 (eEF1A2) is a known proto-oncogene. We proposed that stimulation of the eEF1A2 expression in cancer tissues is caused by the loss of miRNA-mediated control. METHODS: Impact of miRNAs on eEF1A2 at the mRNA and protein levels was examined by qPCR and western blot, respectively. Dual-luciferase assay was applied to examine the influence of miRNAs on 3'-UTR of EEF1A2. To detect miRNA-binding sites, mutations into the 3'-UTR of EEF1A2 mRNA were introduced by the overlap extension PCR. RESULTS: miR-663 and miR-744 inhibited the expression of luciferase gene attached to the 3'-UTR of EEF1A2 up to 20% and 50%, respectively. In MCF7 cells, overexpression of miR-663 and miR-744 reduced the EEF1A2 mRNA level by 30% and 50%. Analogous effects were also observed at the eEF1A2 protein level. In resveratrol-treated MCF7 cells the upregulation of mir-663 and mir-744 was accompanied by downregulation of EEF1A2 mRNA. Both miRNAs were able to inhibit the proliferation of MCF7 cells. CONCLUSION: miR-663 and miR-744 mediate inhibition of the proto-oncogene eEF1A2 expression that results in retardation of the MCF7 cancer cells proliferation. Antitumour effect of resveratrol may include stimulation of the miR-663 and miR-744 expression.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/therapy , MicroRNAs/administration & dosage , Peptide Elongation Factor 1/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/genetics , Cell Movement/genetics , Down-Regulation , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , Peptide Elongation Factor 1/antagonists & inhibitors , Peptide Elongation Factor 1/biosynthesis , Proto-Oncogene Mas , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Resveratrol , Stilbenes/pharmacology , Transfection
4.
Oncogene ; 31(47): 4960-6, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-22266852

ABSTRACT

TWIST1 is a highly conserved basic helix-loop-helix transcription factor that promotes epithelial-mesenchymal transition (EMT). Its misregulation has been observed in various types of tumors. Using the MCF-10A-series of cell lines that recapitulate the early stages of breast cancer formation and EMT, we found TWIST1 to be upregulated during EMT and downregulated early in carcinogenesis. The TWIST1 3'UTR contains putative regulatory elements, including miRNA target sites and two cytoplasmic polyadenylation elements (CPE). We found that miR-580, CPEB1, and CPEB2 act as negative regulators of TWIST1 expression in a sequence-specific and additive/cooperative manner.


Subject(s)
Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Nuclear Proteins/metabolism , Protein Biosynthesis , Twist-Related Protein 1/metabolism , 3' Untranslated Regions , Binding Sites , Breast Neoplasms/pathology , Cell Line , Cell Movement , Disease Progression , Epithelial-Mesenchymal Transition , Female , Genes, Reporter , Humans , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Nuclear Proteins/genetics , RNA Interference , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Twist-Related Protein 1/genetics , Up-Regulation , mRNA Cleavage and Polyadenylation Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/physiology
5.
Oncogene ; 27(30): 4221-32, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18362892

ABSTRACT

Following the screening of a battery of distinct small-interfering RNAs that target various components of the apoptotic machinery, we found that knockdown of the voltage-dependent anion channel 1 (VDAC1) was particularly efficient in preventing cell death induced by cisplatin (CDDP) in non-small cell lung cancer cells. Both the downregulation of VDAC1 and its chemical inhibition with 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid reduced the apoptosis-associated modifications induced by CDDP, including mitochondrial transmembrane potential dissipation and plasma membrane permeabilization. VDAC1 inhibition strongly reduced the CDDP-induced conformational activation of Bax, yet had no discernible effect on the activation of Bak, suggesting that VDAC1 acts downstream of Bak and upstream of Bax. Accordingly, knockdown of Bak abolished the activation of Bax, whereas Bax downregulation had no effect on Bak activation. In VDAC1-depleted cells, the failure of CDDP to activate Bax could be reversed by means of the Bcl-2/Bcl-X(L) antagonist ABT-737, which concomitantly restored CDDP cytotoxicity. Altogether, these results delineate a novel pathway for the induction of mitochondrial membrane permeabilization (MMP) in the course of CDDP-induced cell death that involves a hierarchical contribution of Bak, VDAC1 and Bax. Moreover, our data suggest that VDAC1 may act as a facultative regulator/effector of MMP, depending on the initial cytotoxic event.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Signal Transduction/drug effects , Voltage-Dependent Anion Channel 1/physiology , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Biphenyl Compounds/pharmacology , Cell Death/drug effects , Cell Death/genetics , Cell Membrane Permeability/drug effects , Drug Synergism , HCT116 Cells , HeLa Cells , Humans , Models, Biological , Nitrophenols/pharmacology , Piperazines/pharmacology , Signal Transduction/genetics , Sulfonamides/pharmacology , Tumor Cells, Cultured , Voltage-Dependent Anion Channel 1/antagonists & inhibitors
6.
Biomed Pharmacother ; 60(9): 633-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16962735

ABSTRACT

MicroRNAs (miRNAs) have recently emerged as being essential for development and for the control of cell proliferation/differentiation in various organisms. However, little is known about miRNA function and mode of action at the cellular level. We have designed a miRNA loss-of-function assay, based on chemically modified locked nucleic acids (LNA) antisense oligonucleotides and usable in tissue culture cells. We show that LNA/DNA mixed oligonucleotides form highly stable duplexes with miRNAs in vitro. Ex vivo, the target miRNA becomes undetectable in cells transfected with the antisense oligonucleotide. The effect is dose-dependent, long-lasting, and specific. Moreover, using a reporter assay, we show that antisense LNA/DNA oligonucleotides inhibit short non-coding RNAs at the functional level. Thus LNA/DNA mixmers represent powerful tools for functional analysis of miRNAs.


Subject(s)
MicroRNAs/physiology , Oligonucleotides, Antisense/pharmacology , Cells, Cultured , Humans , MicroRNAs/antagonists & inhibitors , Oligonucleotides
7.
EMBO J ; 20(23): 6816-25, 2001 Dec 03.
Article in English | MEDLINE | ID: mdl-11726517

ABSTRACT

Terminal differentiation of muscle cells follows a precisely orchestrated program of transcriptional regulatory events at the promoters of both muscle-specific and ubiquitous genes. Two distinct families of transcriptional co-activators, GCN5/PCAF and CREB-binding protein (CBP)/p300, are crucial to this process. While both possess histone acetyl-transferase (HAT) activity, previous studies have failed to identify a requirement for CBP/p300 HAT function in myogenic differentiation. We have addressed this issue directly using a chemical inhibitor of CBP/p300 in addition to a negative transdominant mutant. Our results clearly demonstrate that CBP/p300 HAT activity is critical for myogenic terminal differentiation. Furthermore, this requirement is restricted to a subset of events in the differentiation program: cell fusion and specific gene expression. These data help to define the requirements for enzymatic function of distinct coactivators at different stages of the muscle cell differentiation program.


Subject(s)
Acetyltransferases/metabolism , Muscles/physiology , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Animals , Blotting, Western , Cell Differentiation , Cell Line , Chromatin/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Dose-Response Relationship, Drug , E1A-Associated p300 Protein , Genes, Dominant , Genes, Reporter , Histone Acetyltransferases , Immunohistochemistry , Mice , Microscopy, Fluorescence , Mutation , Myogenin/metabolism , Precipitin Tests , Promoter Regions, Genetic , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic , Transcriptional Activation , Transfection
8.
Mol Cell Biol ; 21(20): 7089-96, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564891

ABSTRACT

The protein kinase ribosomal S6 kinase 2 (RSK2) has been implicated in phosphorylation of transcription factor CREB and histone H3 in response to mitogenic stimulation by epidermal growth factor. Binding of phospho-CREB to the coactivator CBP allows gene activation through recruitment of the basal transcriptional machinery. Acetylation of H3 by histone acetyltransferase (HAT) activities, such as the one carried by CBP, has been functionally coupled to H3 phosphorylation. While various lines of evidence indicate that coupled histone acetylation and phosphorylation may act in concert to induce chromatin remodeling events facilitating gene activation, little is known about the coupling of the two processes at the signaling level. Here we show that CBP and RSK2 are associated in a complex in quiescent cells and that they dissociate within a few minutes upon mitogenic stimulus. CBP preferentially interacts with unphosphorylated RSK2 in a complex where both RSK2 kinase activity and CBP acetylase activity are inhibited. Dissociation is dependent on phosphorylation of RSK2 on Ser227 and results in stimulation of both kinase and HAT activities. We propose a model in which dynamic formation and dissociation of the CBP-RSK2 complex in response to mitogenic stimulation allow regulated phosphorylation and acetylation of specific substrates, leading to coordinated modulation of gene expression.


Subject(s)
Acetylesterase/metabolism , Gene Expression Regulation, Enzymologic , Mitogens/metabolism , Nuclear Proteins/metabolism , Phosphotransferases/metabolism , Ribosomal Protein S6 Kinases/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Acetyltransferases/metabolism , Animals , Blotting, Western , COS Cells , CREB-Binding Protein , Epidermal Growth Factor/pharmacology , Glutathione Transferase/metabolism , Histone Acetyltransferases , Humans , Models, Biological , Phorbol Esters/pharmacology , Phosphorylation , Precipitin Tests , Protein Binding , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Transcriptional Activation , Transfection , Ultraviolet Rays
9.
J Biol Chem ; 276(48): 44502-3, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11577095

ABSTRACT

MyoD, an essential transcription factor involved in muscle cell terminal differentiation, is regulated by acetylation, as are a number of other transcription factors, but the histone acetyltransferase enzyme responsible for this acetylation is a matter of controversy. In particular, contradictory findings have been reported concerning the ability of CBP/p300 to acetylate MyoD in vitro. Here we provide an explanation for this discrepancy: although full-length p300 does indeed acetylate MyoD, a fragment of p300 corresponding to its histone acetyltransferase domain does not. In addition to clearly demonstrating that p300 acetylates MyoD in vitro, these results underscore the necessity of using full-length histone acetyltransferase enzymes to draw valid conclusions from acetylation experiments.


Subject(s)
Acetyltransferases/metabolism , MyoD Protein/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Amino Acids/chemistry , HeLa Cells , Histone Acetyltransferases , Humans , Peptides/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/metabolism
10.
EMBO Rep ; 2(9): 794-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11520855

ABSTRACT

The transcription factor E2F, which is a key element in the control of cell proliferation, is repressed by Rb and other pocket proteins in growth-arrested differentiating cells, as well as in proliferating cells when they progress through early G1. It is not known whether similar mechanisms are operative in the two situations. A body of data suggests that E2F repression by pocket proteins involves class I histone deacetylases (HDACs). It has been hypothesized that these enzymes are recruited to E2F target promoters where they deacetylate histones. Here we have tested this hypothesis directly by using formaldehyde cross-linked chromatin immunoprecipitation (XChIP) assays to evaluate HDAC association in living cells. Our data show that a histone deacetylase, HDAC-1, is stably bound to an E2F target promoter during early G1 in proliferating cells and released at the G1-S transition. In addition, our results reveal an inverse correlation between HDAC-1 recruitment and histone H4 acetylation on specific lysines.


Subject(s)
Cell Cycle Proteins , Cell Cycle/physiology , DNA-Binding Proteins , Histone Deacetylases/metabolism , Histones/metabolism , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , 3T3 Cells , Acetylation , Animals , Blotting, Northern , Blotting, Western , Cell Division , Cells, Cultured , Chromatin/metabolism , E2F Transcription Factors , Histone Deacetylase 1 , Lysine/chemistry , Mice , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Time Factors
11.
Mol Cell Biol ; 21(16): 5312-20, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463815

ABSTRACT

Acetylation is emerging as a posttranslational modification of nuclear proteins that is essential to the regulation of transcription and that modifies transcription factor affinity for binding sites on DNA, stability, and/or nuclear localization. Here, we present both in vitro and in vivo evidence that acetylation increases the affinity of myogenic factor MyoD for acetyltransferases CBP and p300. In myogenic cells, the fraction of endogenous MyoD that is acetylated was found associated with CBP or p300. In vitro, the interaction between MyoD and CBP was more resistant to high salt concentrations and was detected with lower doses of MyoD when MyoD was acetylated. Interestingly, an analysis of CBP mutants revealed that the interaction with acetylated MyoD involves the bromodomain of CBP. In live cells, MyoD mutants that cannot be acetylated did not associate with CBP or p300 and were strongly impaired in their ability to cooperate with CBP for transcriptional activation of a muscle creatine kinase-luciferase construct. Taken together, our data suggest a new mechanism for activation of protein function by acetylation and demonstrate for the first time an acetylation-dependent interaction between the bromodomain of CBP and a nonhistone protein.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , MyoD Protein/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Acetylation , Animals , CREB-Binding Protein , Cell Line , Enzyme Activation , Histone Acetyltransferases , Protein Binding , Substrate Specificity , Transcription Factors , p300-CBP Transcription Factors
12.
Cell Mol Life Sci ; 58(5-6): 728-36, 2001 May.
Article in English | MEDLINE | ID: mdl-11437234

ABSTRACT

Differential acetylation of histones and transcription factors plays an important regulatory role in developmental processes, proliferation and differentiation. Aberrant acetylation or deacetylation leads to such diverse disorders as leukemia, epithelial cancers, fragile X syndrome and Rubinstein-Taybi syndrome. The various groups of histone acetyltransferases (CBP/p300, GNAT, MYST, nuclear receptor coactivators and TAFII250) and histone deacetylases are surveyed with regard to their possible or known involvement in cancer progression and human developmental disorders. Current treatment strategies are discussed, which are still mostly limited to histone deacetylase inhibitors such as trichostatin A and butyrate.


Subject(s)
Disease , Histones/chemistry , Histones/metabolism , Saccharomyces cerevisiae Proteins , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Acetylation , Acetyltransferases/metabolism , Animals , Cell Cycle , Cell Differentiation , DNA-Binding Proteins/metabolism , Fragile X Syndrome/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Histone Acetyltransferases , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Trans-Activators/metabolism , Translocation, Genetic/genetics
13.
Oncogene ; 20(24): 3128-33, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11420729

ABSTRACT

The balance between cell differentiation and proliferation is regulated at the transcriptional level. In the cell cycle, the transition from G1 to S phase (G1/S transition) is of paramount importance in this regard. Indeed, it is only before this point that cells can be oriented toward the differentiation pathway: beyond, cells progress into the cycle in an autonomous manner. The G1/S transition is orchestrated by the transcription factor E2F. E2F controls the expression of a group of checkpoint genes whose products are required either for the G1-to-S transition itself or for DNA replication (e.g. DNA polymerase alpha). E2F activity is repressed in growth-arrested cells and in early G1, and is activated at mid-to-late G1. E2F is controlled by the retinoblastoma tumor suppressor protein Rb. Rb represses E2F mainly by recruiting chromatin remodeling factors (histone deacetylases and SWI/SNF complexes), the DNA methyltransferase DNMT1, and a histone methyltransferase. This review will focus on the molecular mechanisms of E2F repression by Rb during the cell cycle and during cell-cycle exit by differentiating cells. A model in which Rb irreversibly represses E2F-regulated genes in differentiated cells by an epigenetic mechanism linked to heterochromatin, and involving histone H3 and promoter DNA methylation, is discussed.


Subject(s)
Carrier Proteins , Chromatin/genetics , DNA-Binding Proteins , Gene Expression Regulation , Retinoblastoma Protein/physiology , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Division , Chromatin/metabolism , E2F Transcription Factors , Humans , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1 , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Biol Chem ; 275(44): 34359-64, 2000 Nov 03.
Article in English | MEDLINE | ID: mdl-10944526

ABSTRACT

The myogenic protein MyoD requires two nuclear histone acetyltransferases, CREB-binding protein (CBP)/p300 and PCAF, to transactivate muscle promoters. MyoD is acetylated by PCAF in vitro, which seems to increase its affinity for DNA. We here show that MyoD is constitutively acetylated in muscle cells. In vitro, MyoD is acetylated both by CBP/p300 and by PCAF on two lysines located at the boundary of the DNA binding domain. MyoD acetylation by CBP/p300 (as well as by PCAF) increases its activity on a muscle-specific promoter, as assessed by microinjection experiments. MyoD mutants that cannot be acetylated in vitro are not activated in the functional assay. Our results provide direct evidence that MyoD acetylation functionally activates the protein and show that both PCAF and CBP/p300 are candidate enzymes for MyoD acetylation in vivo.


Subject(s)
MyoD Protein/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Acetylation , Acetyltransferases/metabolism , Amino Acid Sequence , Animals , E1A-Associated p300 Protein , Histone Acetyltransferases , Mice , Mice, Inbred C3H , Molecular Sequence Data , Transcriptional Activation
15.
Oncogene ; 19(20): 2430-7, 2000 May 11.
Article in English | MEDLINE | ID: mdl-10828885

ABSTRACT

Transforming viral proteins such as E1A which force quiescent cells into S phase have two essential cellular target proteins, Rb and CBP/p300. Rb regulates the G1/S transition by controlling the transcription factor E2F. CBP/p300 is a transcriptional co-activator with intrinsic histone acetyl-transferase activity. This activity is regulated in a cell cycle dependent manner and shows a peak at the G1/S transition, suggesting a function for CBP/p300 in this crucial step of the cell cycle. Here, we have artificially modulated CBP/p300 levels in individual cells through microinjection of specific antibodies and expression vectors. We show that CBP/p300 is required for cell proliferation and has an essential function during the G1/S transition. Using the same microinjection system and GFP-reporter vectors, we demonstrate that CBP/p300 is essential for the activity of E2F, a transcription factor that controls the G1/S transition. In addition, our results suggest that CBP HAT activity is required both for the G1/S transition and for E2F activity. Thus CBP/p300 seems to be a versatile protein involved in opposing cellular processes, which raises the question of how its multiple activities are regulated.


Subject(s)
Acetyltransferases/metabolism , Carrier Proteins , Cell Cycle Proteins/metabolism , G1 Phase , S Phase , 3T3 Cells , Acetyltransferases/genetics , Animals , COS Cells , Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Histone Acetyltransferases , Mice , Mutagenesis , Protein Binding , Retinoblastoma-Binding Protein 1 , Sequence Deletion , TATA-Box Binding Protein , Transcription Factor DP1 , Transcription Factors/metabolism , Transcription, Genetic , p300-CBP Transcription Factors
16.
J Biol Chem ; 275(13): 9797-804, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10734134

ABSTRACT

The retinoblastoma susceptibility gene product, the Rb protein, is a key regulator of mammalian cell proliferation. One of the major targets of Rb is the S phase inducing E2F transcription factor. Once bound to E2F, Rb represses the expression of E2F-regulated genes. Transcriptional repression by Rb is believed to be crucial for the proper control of cell growth. Recently, we and others showed that Rb represses transcription through the recruitment of a histone deacetylase. Interestingly, we show here that the Rb-associated histone deacetylase complex could deacetylate polynucleosomal substrates, indicating that other proteins could be present within this complex. The Rb-associated protein RbAp48 belongs to many histone deacetylase complexes. We show here that the histone deacetylase HDAC1 is able to mediate the formation of a ternary complex containing Rb and RbAp48. Moreover, less deacetylase activity was found associated with Rb in cell extracts depleted for RbAp48 containing complexes, demonstrating that Rb, histone deacetylase, and RbAp48 are physically associated in live cells. Taken together, these data indicate that RbAp48 is a component of the histone deacetylase complex recruited by Rb. Finally, we found that E2F1 and RbAp48 are physically associated in the presence of Rb and HDAC1, suggesting that RbAp48 could be involved in transcriptional repression of E2F-responsive genes.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins , DNA-Binding Proteins , Histone Deacetylases/metabolism , Nuclear Proteins/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma/metabolism , Amino Acid Sequence , Cell Line , E2F Transcription Factors , E2F1 Transcription Factor , G1 Phase , Humans , Molecular Sequence Data , Protein Binding , Retinoblastoma-Binding Protein 1 , Retinoblastoma-Binding Protein 4 , Transcription Factor DP1 , Transcription Factors/metabolism
17.
Proc Natl Acad Sci U S A ; 97(7): 3084-8, 2000 Mar 28.
Article in English | MEDLINE | ID: mdl-10716704

ABSTRACT

Triple-helix-forming oligonucleotides (TFOs), which can potentially modify target genes irreversibly, represent promising tools for antiviral therapies. However, their effectiveness on endogenous genes has yet to be unambiguously demonstrated. To monitor endogenous gene modification by TFOs in a yeast model, we inactivated an auxotrophic marker gene by inserting target sequences of interest into its coding region. The genetically engineered yeast cells then were treated with psoralen-linked TFOs followed by UV irradiation, thus generating highly mutagenic covalent crosslinks at the target site whose repair could restore gene function; the number of revertants and spectrum of mutations generated were quantified. Results showed that a phosphoramidate TFO indeed reaches its target sequence, forms crosslinks, and generates mutations at the expected site via a triplex-mediated mechanism: (i) under identical conditions, no mutations were generated by the same TFO at two other loci in the target strain, nor in an isogenic control strain carrying a modified target sequence incapable of supporting triple-helix formation; (ii) for a given target sequence, whether the triplex was formed in vivo on an endogenous gene or in vitro on an exogenous plasmid, the nature of the mutations generated was identical, and consistent with the repair of a psoralen crosslink at the target site. Although the mutation efficiency was probably too low for therapeutic applications, our results confirm the validity of the triple-helix approach and provide a means of evaluating the effectiveness of new chemically modified TFOs and analogs.


Subject(s)
DNA, Viral/chemistry , Mutation , Nucleic Acid Conformation , Base Sequence , DNA, Viral/genetics , Fungal Proteins/genetics , HIV-1/genetics , HIV-2/genetics , Plasmids
18.
Prog Cell Cycle Res ; 4: 41-7, 2000.
Article in English | MEDLINE | ID: mdl-10740813

ABSTRACT

The critical steps of the cell cycle are generally controlled through the transcriptional regulation of specific subsets of genes. Transcriptional regulation has been recently linked to acetylation or deacetylation of core histone tails: acetylated histone tails are generally associated with active chromatin, whereas deacetylated histone tails are associated with silent parts of the genome. A number of transcriptional co-regulators are histone acetyl-transferases or histone deacetylases. Here, we discuss some of the critical cell cycle steps in which these enzymes are involved.


Subject(s)
Cell Cycle/physiology , Histones/metabolism , Saccharomyces cerevisiae Proteins , Acetylation , Acetyltransferases/metabolism , Animals , Histone Acetyltransferases , Histone Deacetylases/metabolism , Transcription, Genetic
19.
Gene Expr ; 8(1): 33-42, 1999.
Article in English | MEDLINE | ID: mdl-10543729

ABSTRACT

The serum response element (SRE) in the c-fos promoter contains an ets box whose integrity is required for full activation of this proto-oncogene by nerve growth factor (NGF) in PC12 rat pheochromocytoma cells. Electrophoretic mobility shift assays (EMSA) detect a protein in nuclear extracts that binds to the wild-type SRE, but not to an SRE containing a mutated ets box. Competition studies using unlabeled probes, and supershift experiments using antibodies and in vitro translated core serum response factor (SRF) indicate that the protein in question is not YY1, SAP-1, nor Elk-1 and that it does not exhibit ternary complex factor (TCF) activity, so that it may correspond to an autonomously binding Ets family protein. The complete disappearance of this "Ets-like autonomous binding factor" upon terminal differentiation of both L6alpha2 myoblastic and PC12 pheochromocytoma cells points to a possible role in the proliferation/differentiation process.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Response Elements , 3T3 Cells , Animals , Cell Differentiation , Cell Division , Down-Regulation , Mice , Nerve Growth Factor/metabolism , PC12 Cells , Protein Binding , Rats , Serum Response Factor , Tumor Cells, Cultured
20.
Nucleic Acids Res ; 27(20): 3995-4000, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10497263

ABSTRACT

Triplex-forming oligonucleotides (TFOs) are generally designed to inhibit transcription or DNA replication but can be used for more diverse purposes. Here we have designed a chimera peptide-TFO able to activate transcription from a target gene. The designed hybrid molecule contains a triplex-forming sequence, linked through a phosphoroamidate bond to several minimal transcriptional activation domains derived from Herpes simplex virus protein 16 (VP16). We show here that this TFO-peptide chimera (TFO-P) can specifically recognise its DNA target at physiological salt and pH conditions. Bound to the double-stranded target DNA in a promoter region, the TFO-P is able to activate gene expression. Our results suggest that this type of molecule may prove useful in the design of new tools for artificial modulation of gene expression.


Subject(s)
Gene Expression Regulation , Herpes Simplex Virus Protein Vmw65/metabolism , Oligonucleotides/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cell Line , Molecular Sequence Data , Nucleic Acid Conformation , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL