Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters










Publication year range
2.
Proc Natl Acad Sci U S A ; 98(6): 3428-33, 2001 Mar 13.
Article in English | MEDLINE | ID: mdl-11248095

ABSTRACT

Fabry disease is a lipid storage disorder resulting from mutations in the gene encoding the enzyme alpha-galactosidase A (alpha-gal A; EC ). We previously have demonstrated long-term alpha-gal A enzyme correction and lipid reduction mediated by therapeutic ex vivo transduction and transplantation of hematopoietic cells in a mouse model of Fabry disease. We now report marked improvement in the efficiency of this gene-therapy approach. For this study we used a novel bicistronic retroviral vector that engineers expression of both the therapeutic alpha-gal A gene and the human IL-2Ralpha chain (huCD25) gene as a selectable marker. Coexpression of huCD25 allowed selective immunoenrichment (preselection) of a variety of transduced human and murine cells, resulting in enhanced intracellular and secreted alpha-gal A enzyme activities. Of particular significance for clinical applicability, mobilized CD34(+) peripheral blood hematopoietic stem/progenitor cells from Fabry patients have low-background huCD25 expression and could be enriched effectively after ex vivo transduction, resulting in increased alpha-gal A activity. We evaluated effects of preselection in the mouse model of Fabry disease. Preselection of transduced Fabry mouse bone marrow cells elevated the level of multilineage gene-corrected hematopoietic cells in the circulation of transplanted animals and improved in vivo enzymatic activity levels in plasma and organs for more than 6 months after both primary and secondary transplantation. These studies demonstrate the potential of using a huCD25-based preselection strategy to enhance the clinical utility of ex vivo hematopoietic stem/progenitor cell gene therapy of Fabry disease and other disorders.


Subject(s)
Fabry Disease/therapy , Genetic Therapy/methods , Receptors, Interleukin-2/genetics , alpha-Galactosidase/genetics , 3T3 Cells , Animals , Bone Marrow Transplantation , Disease Models, Animal , Fabry Disease/metabolism , Fabry Disease/pathology , Gene Expression , Genetic Vectors , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, Interleukin-2/metabolism , Retroviridae/genetics , alpha-Galactosidase/metabolism
3.
J Biol Chem ; 276(9): 6817-24, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11102443

ABSTRACT

Human histone deacetylases I (HDAC1) and II (HDAC2) are homologous proteins (84% identity) that catalyze release of acetyl groups from modified N-terminal lysines of core histones. Histone deacetylation is correlated with both transient and persistent states of transcriptional inactivity (i.e. silencing) in many eukaryotes. In this study, we analyzed complexes containing HDAC1 and HDAC2 to identify the proteins most stably associated with these deacetylases. Complex cI (9.5 S) contained transcriptional corepressor CoREST/kiaa0071 and a protein homologous to FAD-dependent oxidoreductases, kiaa0601. Complex cII (15 S) contained >/=15 proteins, including CHD3/4 (Mi-2), Mta-L1, RbAp48/46, and MBD3, characteristic of vertebrate nucleosome-remodeling complexes. Under native conditions, cI and cII may contain HDAC1, HDAC2 or both; these can be dissociated to cI and cII core complexes containing only HDAC1 or HDAC2. The (m)CpG-binding protein MBD2 was associated only with the HDAC1 cII core complex. A model is proposed in which HDAC1 core complexes can be targeted to methylated DNA via MBD2 with recruitment of HDAC2 occurring through formation of HDAC1/2 cII dimers. We note that the cI component CoREST/kiaa0071 and the cII component Mta-L1 share a region of homology that includes a SANT domain; this domain may play a role in complex assembly.


Subject(s)
DNA-Binding Proteins , Histone Deacetylases/chemistry , Nerve Tissue Proteins/chemistry , Repressor Proteins/chemistry , Amino Acid Sequence , Co-Repressor Proteins , Conserved Sequence , HeLa Cells , Histone Deacetylases/isolation & purification , Humans , Isoenzymes/chemistry , Molecular Sequence Data , Nerve Tissue Proteins/analysis , Proteins/chemistry , Repressor Proteins/analysis , Riboflavin/analysis , Trans-Activators
4.
EMBO J ; 18(19): 5389-98, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10508171

ABSTRACT

We have investigated ligand-dependent negative regulation of the thyroid-stimulating hormone beta (TSHbeta) gene. Thyroid hormone (T3) markedly repressed activity of the TSHbeta promoter that had been stably integrated into GH(3 )pituitary cells, through the conserved negative regulatory element (NRE) in the promoter. By DNA affinity binding assay, we show that the NRE constitutively binds to the histone deacetylase 1 (HDAC1) present in GH(3 )cells. Significantly, upon addition of T3, the NRE further recruited the thyroid hormone receptor (TRbeta) and another deacetylase, HDAC2. This recruitment coincided with an alteration of in vivo chromatin structure, as revealed by changes in restriction site accessibility. Supporting the direct interaction between TR and HDAC, in vitro assays showed that TR, through its DNA binding domain, strongly bound to HDAC2. Consistent with the role for HDACs in negative regulation, an inhibitor of the enzymes, trichostatin A, attenuated T3-dependent promoter repression. We suggest that ligand-dependent histone deacetylase recruitment is a mechanism of the negative-feedback regulation, a critical function of the pituitary-thyroid axis.


Subject(s)
Feedback , Histone Deacetylases/metabolism , Thyrotropin/genetics , Base Sequence , Chromatin/chemistry , Cyclic AMP/pharmacology , DNA , Gene Expression Regulation/drug effects , Genes, Reporter , Ligands , Protein Binding , Regulatory Sequences, Nucleic Acid , Sequence Deletion , Sequence Homology, Nucleic Acid , Thyrotropin/metabolism , Triiodothyronine/pharmacology
5.
J Biol Chem ; 274(12): 7936-40, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075689

ABSTRACT

Human diploid fibroblasts eventually lose the capacity to replicate in culture and enter a viable but nonproliferative state of senescence. Recently, it has been demonstrated that retroviral-mediated gene transfer into primary fibroblasts of an activated ras gene (V12ras) rapidly accelerates development of the senescent phenotype. Using this in vitro system, we have sought to define the mediators of Ras-induced senescence. We demonstrate that expression of V12Ras results in an increase in intracellular and in particular, mitochondrial reactive oxygen species. The ability of V12Ras to induce growth arrest and senescence is shown to be partially inhibited by coexpression of an activated rac1 gene. A more dramatic rescue of V12Ras-expressing cells is demonstrated when the cells are placed in a low oxygen environment, a condition in which reactive oxygen species production is inhibited. In addition, in a 1% oxygen environment, Ras is unable to trigger an increase in the level of the cyclin-dependent kinase inhibitor p21 or to activate the senescent program. Under normoxic (20% O2) conditions, the V12Ras senescent phenotype is demonstrated to be unaffected by scavengers of superoxide but rescued by scavengers of hydrogen peroxide. These results suggest that in normal diploid cells, Ras proteins regulate oxidant production and that a rise in intracellular H2O2 represents a critical signal mediating replicative senescence.


Subject(s)
Apoptosis/physiology , Reactive Oxygen Species/metabolism , ras Proteins/physiology , Cells, Cultured , Fibroblasts/metabolism , Flow Cytometry , Humans , Oxidation-Reduction , Oxygen/metabolism
6.
J Biol Chem ; 273(38): 24898-905, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733796

ABSTRACT

A unique aspect of the retrovirus life cycle is the obligatory integration of the provirus into host cell chromosomes. Unlike viruses that do not integrate, retroviruses must conserve an ability to activate transcription from a chromatin context. Human immunodeficiency virus (HIV)-1 encodes an unusual and an unusually potent transcriptional transactivator, Tat, which binds to a nascent viral leader RNA, TAR. The action of Tat has been well studied in various reductive model systems; however, the physiological mechanism through which Tat gains access to chromatin-associated proviral long terminal repeats (LTRs) is not understood. We show here that a nuclear histone acetyltransferase activity associates with Tat. Intracellularly, we found that Tat forms a ternary complex with p300 and P/CAF, two histone acetyltransferases (HATs). A murine cell defect in Tat transactivation of the HIV-1 LTR was linked to the reduced abundance of p300 and P/CAF. Thus, overexpression of p300 and P/CAF reconstituted Tat transactivation of the HIV-1 LTR in NIH3T3 cells to a level similar to that observed for human cells. By using transdominant p300 or P/CAF mutants that lack enzymatic activity, we delineated a requirement for the HAT component from the latter but not the former in Tat function. Finally, we observed that Tat-associated HAT is preferentially important for transactivation of integrated, but not unintegrated, HIV-1 LTR.


Subject(s)
Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Gene Products, tat/metabolism , HIV-1/genetics , Proviruses/genetics , Saccharomyces cerevisiae Proteins , Virus Integration , 3T3 Cells , Animals , Binding Sites , Chromatin/genetics , Chromatin/physiology , Cloning, Molecular , Gene Products, tat/genetics , Gene Products, tat/isolation & purification , HIV Long Terminal Repeat , HIV-1/enzymology , HIV-1/physiology , HeLa Cells , Histone Acetyltransferases , Humans , Life Cycle Stages , Mice , Proviruses/physiology , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Transcription Factors , p300-CBP Transcription Factors , tat Gene Products, Human Immunodeficiency Virus
7.
Cell ; 94(1): 35-44, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9674425

ABSTRACT

PCAF histone acetylase plays a role in regulation of transcription, cell cycle progression, and differentiation. Here, we show that PCAF is found in a complex consisting of more than 20 distinct polypeptides. Strikingly, some polypeptides are identical to TBP-associated factors (TAFs), which are subunits of TFIID. Like TFIID, histone fold-containing factors are present within the PCAF complex. The histone H3- and H2B-like subunits within the PCAF complex are identical to those within TFIID, namely, hTAF(II)31 and hTAF(II)20/15, respectively. The PCAF complex has a novel histone H4-like subunit with similarity to hTAF(II)80 that interacts with the histone H3-like domain of hTAF(II)31. Moreover, the PCAF complex has a novel subunit with WD40 repeats having a similarity to hTAF(II)100.


Subject(s)
Acetyltransferases/chemistry , DNA-Binding Proteins/isolation & purification , Histones/isolation & purification , Nuclear Proteins/isolation & purification , Saccharomyces cerevisiae Proteins , Transcription Factors/isolation & purification , Amino Acid Sequence , Fungal Proteins , HeLa Cells , Histone Acetyltransferases , Humans , Macromolecular Substances , Mass Spectrometry , Molecular Sequence Data , Protein Kinases , Sequence Analysis , Sequence Homology, Amino Acid
8.
Virology ; 243(2): 331-9, 1998 Apr 10.
Article in English | MEDLINE | ID: mdl-9568032

ABSTRACT

Components of the eukaryotic vaccinia virus/T7 RNA polymerase hybrid expression system were assessed using recombinant and nonrecombinant forms of modified vaccinia Ankara (MVA), a replication-deficient vaccinia virus strain. Recombinant MVA virus expressing T7 RNA polymerase (Wyatt, L. S., Moss, B., and Rozenblatt, S. (1995). Virology 210, 202-205) stimulated high levels of expression from a T7 promoter-chloramphenicol acetyltransferase (CAT) reporter. Most, but not all, of the virally induced expression was T7 RNA polymerase and T7 promoter dependent, with no viral enhancement of translation of T7 transcripts. The efficacy of supplying T7 RNA polymerase expression from nonviral sources was evaluated using a self-amplifying T7 RNA polymerase autogene or an inducible T7 RNA polymerase expression vector. The latter modes yielded CAT activity dependent on T7 RNA polymerase expression; however, expression required viral factors independent of T7 RNA polymerase and did not reach that attained using the recombinant virus. In further experiments, MVA-induced T7 RNA polymerase expression was upregulated by alpha-amanitin, an inhibitor of eukaryotic polymerases. This indicates that MVA/T7 RNA polymerase hybrid expression may be rendered still more efficient by ameliorating transcriptional interference due to an alpha-amanitin-sensitive eukaryotic factor(s).


Subject(s)
Amanitins/metabolism , DNA-Directed RNA Polymerases/genetics , Defective Viruses/enzymology , Gene Expression , Genetic Vectors , Vaccinia virus/enzymology , Amanitins/genetics , Chloramphenicol O-Acetyltransferase/genetics , DNA-Directed RNA Polymerases/metabolism , Defective Viruses/genetics , Defective Viruses/physiology , Enzyme Induction , Enzyme Inhibitors/metabolism , HeLa Cells , Humans , Promoter Regions, Genetic , Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Proteins , Virus Replication
10.
Mutat Res ; 385(1): 31-9, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9372846

ABSTRACT

A new approach to monitoring UV damage and repair in the human genome has been developed. The proposed approach is based on a combination of features unique to interspersed repetitive Alu elements, and the ability of certain DNA lesions to block Taq polymerase-mediated DNA synthesis: namely, the extraordinary abundance of Alu repeats throughout the human genome in conjunction with distinct sequence motifs comprising long runs of T residues which are likely targets for formation of UV lesions. Hence, Taq polymerase-mediated extension synthesis with Alu specific primers was employed to visualize formation of discrete predicted adducts within the element. Several variations of the Alu-primer driven amplification protocol were developed to monitor the following aspects of damage: (i) induction of UV-photoproducts at predicted sites within the Alu sequence, (ii) modification of extension synthesis profiles, (iii) UV dose dependent, quantitative inhibition of Alu-primer driven amplification. The assays reveal sites of predicted Taq polymerase blockage within the Alu sequence, a global decrease in the mean length of extension products, and a measurable reduction in the quantity of extension products that is inversely proportional to UV dose. Thus, the exceptional abundance of Alu repeats and their primary sequence features, in combination with the ability of UV lesions to block elongation by Taq polymerase, provide a novel and sensitive system for detecting UV damage in the human genome. The system detects UV damage at levels that are compatible with cellular DNA repair, and provides a unique amplification-based protocol for probing the overall integrity of human DNA.


Subject(s)
DNA Damage , Polymerase Chain Reaction/methods , Repetitive Sequences, Nucleic Acid , Cell Line , DNA/radiation effects , DNA Adducts/analysis , Fibroblasts , Genome , Humans , Ultraviolet Rays
11.
Proc Natl Acad Sci U S A ; 94(21): 11295-300, 1997 Oct 14.
Article in English | MEDLINE | ID: mdl-9326603

ABSTRACT

Histone acetylation is thought to have a role in transcription. To gain insight into the role of histone acetylation in retinoid-dependent transcription, we studied the effects of trichostatin A (TSA), a specific inhibitor of histone deacetylase, on P19 embryonal carcinoma cells. We show that coaddition of TSA and retinoic acid (RA) markedly enhances neuronal differentiation in these cells, although TSA alone does not induce differentiation but causes extensive apoptosis. Consistent with the cooperative effect of TSA and RA, coaddition of the two agents synergistically enhanced transcription from stably integrated RA-responsive promoters. The transcriptional synergy by TSA and RA required the RA-responsive element and a functional retinoid X receptor (RXR)/retinoic acid receptor (RAR) heterodimer, both obligatory for RA-dependent transcription. Furthermore, TSA led to promoter activation by an RXR-selective ligand that was otherwise inactive in transcription. In addition, TSA enhanced transcription from a minimum basal promoter, independently of the RA-responsive element. Finally, we show that TSA alone or in combination with RA increases in vivo endonuclease sensitivity within the RA-responsive promoter, suggesting that TSA treatment might alter a local chromatin environment to enhance RXR/RAR heterodimer action. Thus, these results indicate that histone acetylation influences activity of the heterodimer, which is in line with the observed interaction between the RXR/RAR heterodimer and a histone acetylase presented elsewhere.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Receptors, Retinoic Acid/physiology , Transcription Factors/physiology , Tretinoin/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Embryonal , Cell Cycle/drug effects , Cell Differentiation/drug effects , Dimerization , Genes, Reporter , Kinetics , Luciferases/biosynthesis , Mice , Neurons/cytology , Promoter Regions, Genetic , Receptors, Retinoic Acid/biosynthesis , Recombinant Proteins/biosynthesis , Retinoid X Receptors , Time Factors , Transcription Factors/biosynthesis , Transfection , Tumor Cells, Cultured
12.
Mol Cell Biol ; 17(8): 4877-82, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9234744

ABSTRACT

The p21(WAF1/CIP1/sdi1) gene product (WAF1) inhibits DNA replication in vitro (J. Chen, P. Jackson, M. Kirschner, and A. Dutta, Nature 374:386-388, 1995; S. Waga, G. Hannon, D. Beach, and B. Stillman, Nature 369:574-578, 1994), but in vivo studies on the antiproliferative activity of WAF1 have not resolved G1-phase arrest from potential inhibition of S-phase progression. Here, we demonstrate that elevated WAF1 expression can retard replicative DNA synthesis in vivo. The WAF1-mediated inhibitory effect could be antagonized by cyclin A, cyclin E, or the simian virus 40 small-t antigen with no decrease in the levels of WAF1 protein in transfected cells. Proliferating-cell nuclear antigen (PCNA) overexpression was neither necessary nor sufficient to antagonize WAF1 action. Expression of the N-terminal domain of WAF1, responsible for cyclin-dependent kinase (CDK) interaction, had the same effect as full-length WAF1, while the PCNA binding C terminus exhibited modest activity. We conclude that S-phase progression in mammalian cells is dependent on continuing cyclin and CDK activity and that WAF1 affects S phase primarily through cyclin- and CDK-dependent pathways.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/physiology , Proto-Oncogene Proteins , S Phase/physiology , Antigens, Polyomavirus Transforming/genetics , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/physiology , Cyclins/genetics , DNA Replication , Enzyme Inhibitors , G1 Phase/physiology , Gene Expression , Humans , Osteosarcoma , Phosphoprotein Phosphatases/metabolism , Proliferating Cell Nuclear Antigen/physiology , Protein Serine-Threonine Kinases/physiology , Simian virus 40/immunology , Transfection , Tumor Cells, Cultured
13.
Biochemistry ; 36(31): 9493-500, 1997 Aug 05.
Article in English | MEDLINE | ID: mdl-9235994

ABSTRACT

We have analyzed the effects on HeLa cells of reduction of the CAS protein, the human homologue to yeast chromosome segregation protein CSE1. Expression of CAS antisense cDNA decreases the amount of CAS protein in HeLa cells and perturbs progression from G2 (retards transition from G2) to G1 in the cell cycle. Increased levels of cyclin B in CAS antisense transfected cells correlated with an arrest in G2 phase or mitosis. This arrest upon CAS attenuation is consistent with observations that yeast with CSE1 mutations are defective in mitosis and cyclin B degradation.


Subject(s)
DNA, Antisense/pharmacology , G1 Phase/genetics , G2 Phase/genetics , Proteins/antagonists & inhibitors , Cellular Apoptosis Susceptibility Protein , Cyclins/metabolism , DNA, Complementary , Electroporation , Flow Cytometry , HeLa Cells , Humans , Transfection
14.
Mol Cell ; 1(1): 35-45, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9659901

ABSTRACT

PCAF is a histone acetyltransferase that associates with p300/CBP and competes with E1A for access to them. While exogenous expression of PCAF potentiates both MyoD-directed transcription and myogenic differentiation, PCAF inactivation by anti-PCAF antibody microinjection prevents differentiation. MyoD interacts directly with both p300/CBP and PCAF, forming a multimeric protein complex on the promoter elements. Viral transforming factors that interfere with muscle differentiation disrupt this complex without affecting the MyoD-DNA interaction, indicating functional significance of the complex formation. Exogenous expression of PCAF or p300 promotes p21 expression and terminal cell-cycle arrest. Both of these activities are dependent on the histone acetyltransferase activity of PCAF, but not on that of p300. These results indicate that recruitment of histone acetyltransferase activity of PCAF by MyoD, through p300/CBP, is crucial for activation of the myogenic program.


Subject(s)
Acetyl-CoA C-Acyltransferase/metabolism , Acetyltransferases/metabolism , Cell Cycle Proteins/metabolism , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/cytology , Saccharomyces cerevisiae Proteins , Trans-Activators , Animals , Antigens, Viral, Tumor/pharmacology , CREB-Binding Protein , Cell Differentiation/physiology , Cells, Cultured , E1A-Associated p300 Protein , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Histone Acetyltransferases , Mice , Multienzyme Complexes/metabolism , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/chemistry , Muscle, Skeletal/enzymology , MyoD Protein/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacology , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Transcription Factors/pharmacology , Transcriptional Activation/physiology , p300-CBP Transcription Factors
15.
J Immunol ; 157(11): 5145-54, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8943426

ABSTRACT

Type I (alpha,beta) and type II (gamma) IFNs elicit antiproliferative and antiviral activities through two distinct transcription pathways involving 1) IRF family proteins and ISGF3, and 2) STAT1. We have employed a dominant negative strategy to study the role of IRF family proteins in eliciting the biologic activities of IFN. A truncated IRF protein retaining the DNA-binding domain (DBD) of ICSBP (a member of the IRF family) was stably transfected into U937 monocytic cells. Clones expressing DBD had markedly reduced ISRE-binding activity and were defective in expressing several type I IFN-inducible genes. STAT1 was one such type I IFN-inducible gene whose expression was also inhibited in DBD clones. As a result, the expression of several IFN-gamma-inducible genes was also inhibited in these clones, indicating functional coupling of the type I and type II IFN transcription pathways. Furthermore, DBD clones grew more slowly than control clones and were refractory to antiproliferative effects of both types of IFNs. We found that IFN treatment of U937 cells leads to a G1 arrest and an increase in underphosphorylated retinoblastoma gene product. However, IFN treatment did not change the cell cycle profile, nor retinoblastoma gene product phosphorylation state in DBD clones. These data indicate that expression of DBD disrupts cell cycle regulatory mechanisms. Combined with the previously noted failure of DBD clones to elicit antiviral activity, the present work shows that IRF family proteins play an integral part in growth control activities of IFNs.


Subject(s)
DNA-Binding Proteins/genetics , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Mutation , Repressor Proteins , Transcription Factors/genetics , Animals , Antibodies/pharmacology , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Division/drug effects , Cell Division/genetics , Cell Line , Clone Cells , DNA-Binding Proteins/physiology , Gene Expression/drug effects , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Interferon Regulatory Factors , Interferon-Stimulated Gene Factor 3 , Interferon-Stimulated Gene Factor 3, gamma Subunit , Phosphoproteins/genetics , Phosphoproteins/physiology , Phosphorylation , Rabbits , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , STAT1 Transcription Factor , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors/physiology , Transfection
16.
Cell ; 87(5): 953-9, 1996 Nov 29.
Article in English | MEDLINE | ID: mdl-8945521

ABSTRACT

p300/CBP is a transcriptional adaptor that integrates signals from many sequence-specific activators via direct interactions. Various cellular and viral factors target p300/CBP to modulate transcription and/or cell cycle progression. One such factor, the cellular p300/CBP associated factor (PCAF), possesses intrinsic histone acetyltransferase activity. Here, we demonstrate that p300/CBP is not only a transcriptional adaptor but also a histone acetyltransferase. p300/CBP represents a novel class of acetyltransferases in that it does not have the conserved motif found among various other acetyltransferases. p300/CBP acetylates all four core histones in nucleosomes. These observations suggest that p300/CBP acetylates nucleosomes in concert with PCAF.


Subject(s)
Acetyltransferases/metabolism , Histones/metabolism , Nuclear Proteins/metabolism , Trans-Activators , Transcription Factors/metabolism , Transcription, Genetic/physiology , Acetylation , Adenoviridae/enzymology , Amino Acid Sequence , CREB-Binding Protein , Histones/chemistry , Lysine/metabolism , Molecular Sequence Data , Nuclear Proteins/chemistry , Nucleosomes/chemistry , Nucleosomes/enzymology , Phosphorylation , Protein Structure, Tertiary , Transcription Factors/chemistry , Viral Proteins/metabolism
17.
Mol Cell Biol ; 16(9): 5210-8, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8756678

ABSTRACT

Human diploid fibroblasts (HDF) complete a limited number of cell divisions before entering a growth arrest state that is termed replicative senescence. Two histone deacetylase inhibitors, sodium butyrate and trichostatin A, dramatically reduce the HDF proliferative life span in a manner that is dependent on one or more cell doublings in the presence of these agents. Cells arrested and subsequently released from histone deacetylase inhibitors display markers of senescence and exhibit a persistent G1 block but remain competent to initiate a round of DNA synthesis in response to simian virus 40 T antigen. Average telomere length in prematurely arrested cells is greater than in senescent cells, reflecting a lower number of population doublings completed by the former. Taken together, these results support the view that one component of HDF senescence mimics a cell cycle-dependent drift in differentiation state and that propagation of HDF in histone deacetylase inhibitors accentuates this component.


Subject(s)
Butyrates/pharmacology , Cell Cycle/physiology , Cellular Senescence/drug effects , Enzyme Inhibitors/pharmacology , Fibroblasts/drug effects , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/physiology , Butyric Acid , Cell Division , Cell Line , Cellular Senescence/physiology , Fibroblasts/chemistry , Fibroblasts/cytology , HeLa Cells/drug effects , Histone Deacetylases/physiology , Humans , Retinoblastoma Protein/physiology , Telomere/ultrastructure , Transfection
18.
FASEB J ; 10(9): 1071-7, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8801169

ABSTRACT

Retinoids cause differentiation in embryonal carcinoma (EC) cells, thus mimicking events in mammalian development. Here, we show that retinoids also cause apoptosis in P19 EC cells. Characteristic DNA fragmentation was observed within 36 h after addition of retinoic acid (RA). Synthetic retinoids that are selective for RA receptors (RAR) were also effective in inducing apoptosis, whereas RXR selective ligands were without effect. The combination of RAR and RXR ligands resulted in a synergistic increase in apoptotic cell death. As with apoptosis, neuronal differentiation of P19 cells was synergistically induced by the combination of RAR and RXR ligands. Data obtained with an RAR antagonist and with P19 cells carrying a dominant negative RXR indicate that the two processes are receptor mediated. Together, our results indicate that retinoid-induced apoptosis and neuronal differentiation are closely coupled, and that both RAR and RXR play a role in these processes as active receptors for their respective ligands.


Subject(s)
Apoptosis , Neurons/cytology , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Carcinoma, Embryonal , Cell Adhesion , Cell Cycle/drug effects , Cell Differentiation , DNA Damage , Dose-Response Relationship, Drug , Drug Synergism , Ligands , Retinoid X Receptors , Transcription Factors , Tumor Cells, Cultured
19.
Nature ; 382(6589): 319-24, 1996 Jul 25.
Article in English | MEDLINE | ID: mdl-8684459

ABSTRACT

The adenoviral oncoprotein E1A induces progression through the cell cycle by binding to the products of the p300/CBP and retinoblastoma gene families. A new cellular p300/CBP-associated factor (P/CAF) having intrinsic histone acetylase activity has been identified that competes with E1A. Exogenous expression of P/CAF in HeLa cells inhibits cell-cycle progression and counteracts the mitogenic activity of E1A. E1A disturbs the normal cellular interaction between p300/CBP and its associated histone acetylase.


Subject(s)
Adenovirus E1A Proteins/metabolism , Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators , Transcription Factors/metabolism , Acetyltransferases/genetics , Acetyltransferases/metabolism , Amino Acid Sequence , Binding, Competitive , Cell Cycle , Cell Cycle Proteins/genetics , Cloning, Molecular , Escherichia coli , HeLa Cells , Histone Acetyltransferases , Humans , Molecular Sequence Data , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , p300-CBP Transcription Factors
20.
J Biol Chem ; 271(10): 5819-23, 1996 Mar 08.
Article in English | MEDLINE | ID: mdl-8621451

ABSTRACT

The nature of chromatin organization over Alu repetitive elements is of interest with respect to the maintenance of their transcriptional silencing as well as their potential to influence local chromatin structure. We previously demonstrated that the pattern of nucleosomal organization over Alu elements in native chromatin is specific and similar to the pattern observed with an in vitro reconstituted Alu template. This pattern, distinguished by a nucleosome centered over the 5 -end of the Alu element, is associated with repression of polymerase III-dependent transcription in vitro (Englander, E. W., Wolffe, A. P., and Howard, B. H. (1993) J. Biol. Chem. 268, 19565-19573; Englander, E. W., and Howard, B. H. (1995) J. Biol. Chem. 270, 10091-10096). In the current study, additional templates representing both evolutionarily old and young Alu subfamilies were found to direct a similar pattern of nucleosome assembly, consistent with the view that nucleosome positioning in vitro is shared by a majority of Alus. We discovered however, that the specific nucleosome positioning pattern was disrupted over one member of a young Alu subfamily, which recently transposed immediately downstream to a T14A11 sequence in the neurofibromatosis type 1 locus (Wallace, M. R., Andersen, L. B., Saulino, A. M., Gregory, P. E., Glover, T. W., and Collins, F. S. (1991) Nature 353, 864-866). Upon removal of this sequence motif, the expected pattern of assembly was restored to the neurofibromatosis type 1-Alu template. This finding indicates that, at least in vitro, certain sequences can override the propensity for positioning nucleosomes that is inherent to Alu elements. The finding also raises the possibility that a similar situation may occur in vivo, with potential implications for understanding mechanisms by which certain Alu elements may evade chromatin-mediated transcriptional silencing.


Subject(s)
Genes, Neurofibromatosis 1 , Nucleosomes/physiology , Protein Biosynthesis , Proteins/genetics , Repetitive Sequences, Nucleic Acid , Animals , Biological Evolution , Chickens , Chromatin/physiology , Cloning, Molecular , Cytidine Triphosphate/metabolism , DNA Footprinting , Deoxyribonuclease I , Erythrocytes/metabolism , Histones/isolation & purification , Histones/metabolism , Humans , Multigene Family , Neurofibromin 1 , Restriction Mapping , Templates, Genetic , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...