Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
1.
J Cell Biochem ; 100(2): 385-401, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-16924674

ABSTRACT

Poly(ADP-ribose) polymerase 3 (PARP-3) is a novel member of the PARP family of enzymes that synthesize poly(ADP-ribose) on themselves and other acceptor proteins. Very little is known about this PARP, which is closely related to PARP-1 and PARP-2. By sequence analysis, we find that PARP-3 may be expressed in two isoforms which we studied in more detail to gain insight into their possible functions. We find that both PARP-3 isoforms, transiently expressed as GFP or FLAG fusions, are nuclear. Detection of endogenous PARP-3 with a specific antibody also shows a widespread nuclear distribution, appearing in numerous small foci and a small number of larger foci. Through co-localization experiments and immunoprecipitations, the larger nuclear foci were identified as Polycomb group bodies (PcG bodies) and we found that PARP-3 is part of Polycomb group protein complexes. Furthermore, using a proteomics approach, we determined that both PARP-3 isoforms are part of complexes comprising DNA-PKcs, PARP-1, DNA ligase III, DNA ligase IV, Ku70, and Ku80. Our findings suggest that PARP-3 is a nuclear protein involved in transcriptional silencing and in the cellular response to DNA damage.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA/genetics , Poly(ADP-ribose) Polymerases/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Antigens, Nuclear/metabolism , Base Sequence , Cell Cycle Proteins/genetics , Cell Line , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Enzymologic , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Ku Autoantigen , Mass Spectrometry , Molecular Sequence Data , Poly(ADP-ribose) Polymerases/genetics , Polycomb-Group Proteins , Protein Binding , Repressor Proteins/genetics
2.
J Theor Biol ; 239(3): 298-312, 2006 Apr 07.
Article in English | MEDLINE | ID: mdl-16162356

ABSTRACT

Splicing factor (SF) compartments, also known as speckles, are heterogeneously distributed compartments within the nucleus of eukaryotic cells that are enriched in pre-mRNA SFs. We derive a fourth-order aggregation-diffusion model that describes a possible mechanism underlying the organization of SFs into speckles. The model incorporates two hypotheses, namely (1) that self-organization of dephosphorylated SFs, modulated by a phosphorylation-dephosphorylation cycle, is responsible for the formation and disappearance of speckles, and (2) that an underlying nuclear structure plays a major role in the organization of SFs. A linear stability analysis about homogeneous steady-state solutions of the model reveals how the self-interaction among dephosphorylated SFs can result in the onset of spatial patterns. A detailed bifurcation analysis of the model describes how phosphorylation and dephosphorylation modulate the onset of the compartmentalization of SFs.


Subject(s)
Cell Compartmentation/physiology , Eukaryotic Cells/ultrastructure , Models, Biological , Nuclear Proteins/metabolism , RNA Splicing , Animals , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Eukaryotic Cells/metabolism , Microscopy, Fluorescence , Phosphorylation , Protein Binding/physiology
3.
Bull Math Biol ; 66(6): 1515-45, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15522344

ABSTRACT

Fluorescence recovery after photobleaching (FRAP) is an experimental technique used to measure the mobility of proteins within the cell nucleus. After proteins of interest are fluorescently tagged for their visualization and monitoring, a small region of the nucleus is photobleached. The experimental FRAP data are obtained by recording the recovery of the fluorescence in this region over time. In this paper, we characterize the fluorescence recovery curves for diffusing nuclear proteins undergoing binding events with an approximate spatially homogeneous structure. We analyze two mathematical models for interpreting the experimental FRAP data, namely a reaction-diffusion model and a compartmental model. Perturbation analysis leads to a clear explanation of two important limiting dynamical types of behavior exhibited by experimental recovery curves, namely, (1) a reduced diffusive recovery, and (2) a biphasic recovery characterized by a fast phase and a slow phase. We show how the two models, describing the same type of dynamics using different approaches, relate and share common ground. The results can be used to interpret experimental FRAP data in terms of protein dynamics and to simplify the task of parameter estimation. Application of the results is demonstrated for nuclear actin and type H1 histone.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Genome , Histones/chemistry , Kinetics , Mathematical Computing , Models, Biological , Nuclear Proteins/genetics , Protein Binding , Spectrometry, Fluorescence/methods
4.
Mol Biol Cell ; 12(10): 3046-59, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11598190

ABSTRACT

DEAD box proteins are putative RNA helicases that function in all aspects of RNA metabolism, including translation, ribosome biogenesis, and pre-mRNA splicing. Because many processes involving RNA metabolism are spatially organized within the cell, we examined the subcellular distribution of a human DEAD box protein, DDX1, to identify possible biological functions. Immunofluorescence labeling of DDX1 demonstrated that in addition to widespread punctate nucleoplasmic labeling, DDX1 is found in discrete nuclear foci approximately 0.5 microm in diameter. Costaining with anti-Sm and anti-promyelocytic leukemia (PML) antibodies indicates that DDX1 foci are frequently located next to Cajal (coiled) bodies and less frequently, to PML bodies. Most importantly, costaining with anti-CstF-64 antibody indicates that DDX1 foci colocalize with cleavage bodies. By microscopic fluorescence resonance energy transfer, we show that labeled DDX1 resides within a Förster distance of 10 nm of labeled CstF-64 protein in both the nucleoplasm and within cleavage bodies. Coimmunoprecipitation analysis indicates that a proportion of CstF-64 protein resides in the same complex as DDX1. These studies are the first to identify a DEAD box protein associating with factors involved in 3'-end cleavage and polyadenylation of pre-mRNAs.


Subject(s)
Cell Nucleus/metabolism , RNA Helicases/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Cell Cycle/physiology , Cells, Cultured , DEAD-box RNA Helicases , Fibroblasts , HeLa Cells , Humans , Mice , Microscopy, Confocal , Precipitin Tests , RNA Helicases/ultrastructure , RNA Precursors/ultrastructure , RNA, Messenger/ultrastructure , RNA-Binding Proteins/ultrastructure , Subcellular Fractions/metabolism , Tumor Cells, Cultured , mRNA Cleavage and Polyadenylation Factors
5.
J Biol Chem ; 276(41): 38307-19, 2001 Oct 12.
Article in English | MEDLINE | ID: mdl-11479283

ABSTRACT

Histone acetylation, a reversible modification of the core histones, is widely accepted to be involved in remodeling chromatin organization for genetic reprogramming. Histone acetylation is a dynamic process that is regulated by two classes of enzymes, the histone acetyltransferases (HATs) and histone deacetylases (HDACs). Although promoter-specific acetylation and deacetylation has received most of the recent attention, it is superimposed upon a broader acting and dynamic acetylation that profoundly affects many nuclear processes. In this study, we monitored this broader histone acetylation as cells enter and exit mitosis. In contrast to the hypothesis that HATs and HDACs remain bound to mitotic chromosomes to provide an epigenetic imprint for postmitotic reactivation of the genome, we observed that HATs and HDACs are spatially reorganized and displaced from condensing chromosomes as cells progress through mitosis. During mitosis, HATs and HDACs are unable to acetylate or deacetylate chromatin in situ despite remaining fully catalytically active when isolated from mitotic cells and assayed in vitro. Our results demonstrate that HATs and HDACs do not stably bind to the genome to function as an epigenetic mechanism of selective postmitotic gene activation. Our results, however, do support a role for spatial organization of these enzymes within the cell nucleus and their relationship to euchromatin and heterochromatin postmitotically in the reactivation of the genome.


Subject(s)
Acetyltransferases/metabolism , Chromatin/metabolism , Histone Deacetylases/metabolism , Mitosis , Saccharomyces cerevisiae Proteins , Acetylation , Animals , Blotting, Western , Cell Line , Histone Acetyltransferases , Microscopy, Fluorescence , Phosphorylation
6.
Biochem Cell Biol ; 79(3): 253-66, 2001.
Article in English | MEDLINE | ID: mdl-11467739

ABSTRACT

The CREB binding protein (CBP) was first identified as a protein that specifically binds to the active phosphorylated form of the cyclic-AMP response element binding protein (CREB). CBP was initially defined as a transcriptional coactivator that, as a result of its large size and multiple protein binding domain modules, may function as a molecular scaffold. More recently, an acetyltransferase activity, both of histones and nonhistones, has been found to be essential for transactivation. In this review, we will discuss the current understanding of the acetyltransferase specificity and activity of the CBP protein and how it may function to coactivate transcription. We will also examine the regulation of the CBP histone acetyltransferase activity in the cell cycle, by signal-transduction pathways and throughout development.


Subject(s)
Acetyltransferases/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Acetyltransferases/genetics , Animals , CREB-Binding Protein , Cell Cycle/physiology , Cell Nucleus/chemistry , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , Hematopoiesis/physiology , Histones/chemistry , Histones/metabolism , Humans , Microscopy, Confocal , Nuclear Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rubinstein-Taybi Syndrome/genetics , Signal Transduction , Trans-Activators/genetics
7.
Biochem Cell Biol ; 79(3): 267-74, 2001.
Article in English | MEDLINE | ID: mdl-11467740

ABSTRACT

Living cells can filter the same set of biochemical signals to produce different functional outcomes depending on the deformation of the cell. It has been suggested that the cell may be "hard-wired" such that external forces can mediate internal nuclear changes through the modification of established, balanced, internal cytoskeletal tensions. This review will discuss the potential of subnuclear structures and nuclear chromatin to participate in or respond to transduction of mechanical signals originating outside the nucleus. The mechanical interactions of intranuclear structure with the nuclear lamina will be examined. The nuclear lamina, in turn, provides a structural link between the nucleus and the cytoplasmic and cortical cytoskeleton. These mechanical couplings may provide a basis for regulating gene expression through changes in cell shape.


Subject(s)
Cell Nucleus/physiology , Chromatin/metabolism , Gene Expression Regulation/physiology , Signal Transduction/physiology , Cell Size , Cytoskeleton/metabolism , Female , Humans , Nucleic Acid Conformation , Stress, Mechanical
8.
J Biol Chem ; 276(38): 35826-35, 2001 Sep 21.
Article in English | MEDLINE | ID: mdl-11466315

ABSTRACT

Histone deacetylases (HDACs) are part of transcriptional corepressor complexes and play key roles in regulating chromatin structure. Three different classes of human HDACs have been defined based on their homology to HDACs found in Saccharomyces cerevisiae: RPD3 (class I), HDA1 (class II), and SIR2 (class III). Here we describe the identification and functional characterization of HDAC7, a new member of the human class II HDAC family. Although HDAC7 is localized mostly to the cell nucleus, it is also found in the cytoplasm, suggesting nucleocytoplasmic shuttling. The HDAC activity of HDAC7 maps to a carboxyl-terminal domain and is dependent on the interaction with the class I HDAC, HDAC3, in the cell nucleus. Cytoplasmic HDAC7 that is not bound to HDAC3 is enzymatically inactive. We provide evidence that the transcriptional corepressors SMRT and N-CoR could serve as critical mediators of HDAC7 activity by binding class II HDACs and HDAC3 by two distinct repressor domains. Different class II HDACs reside in the cell nucleus in stable and autonomous complexes with enzymatic activity, but the enzymatic activities associated with HDAC7 and HDAC4 rely on shared cofactors, including HDAC3 and SMRT/N-CoR.


Subject(s)
Histone Deacetylases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Nucleus/enzymology , Cytoplasm/enzymology , DNA Primers , Histone Deacetylases/chemistry , Humans , Molecular Sequence Data , Saccharomyces cerevisiae/enzymology , Sequence Homology, Amino Acid
9.
J Steroid Biochem Mol Biol ; 76(1-5): 9-21, 2001.
Article in English | MEDLINE | ID: mdl-11384859

ABSTRACT

The cell nucleus is increasingly recognized as a spatially organized structure. In this review, the nature and controversies associated with nuclear compartmentalization are discussed. The relationship between nuclear structure and organization of proteins involved in the regulation of RNA polymerase II-transcribed genes is then discussed. Finally, very recent data on the mobility of these proteins within the cell nucleus is considered and their implications for regulation through compartmentalization of proteins and genomic DNA are discussed.


Subject(s)
Cell Compartmentation , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Acetylation , Chromatin/chemistry , Chromatin/metabolism , Histones/metabolism , Humans , Interphase , Protein Conformation , Receptors, Estrogen/metabolism , Transcription Factors/metabolism
10.
J Cell Biol ; 152(5): 1099-106, 2001 Mar 05.
Article in English | MEDLINE | ID: mdl-11238464

ABSTRACT

The transcription coactivator and histone acetyltransferase CAMP response element-binding protein (CBP) has been demonstrated to accumulate in promyelocytic leukemia (PML) bodies. We show that this accumulation is cell type specific. In cells where CBP does not normally accumulate in PML bodies, it can be induced to accumulate in PML bodies through overexpression of either CBP or Pml, but not Sp100. Using fluorescence recovery after photobleaching, we demonstrate that CBP moves rapidly into and out of PML bodies. In contrast, Pml and Sp100 are relatively immobile in the nucleoplasm and within PML nuclear bodies. They possess the characteristics expected of proteins that would play a structural role in the integrity of these subnuclear domains. Our results are consistent with CBP being a dynamic component of PML bodies and that the steady-state level in these structures can be modulated by Pml.


Subject(s)
Antigens, Nuclear , Cell Nucleus Structures/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/drug effects , Fluorescence , Fluorescent Antibody Technique , Humans , Interferons/pharmacology , Leukemia, Promyelocytic, Acute/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Matrix/chemistry , Nuclear Matrix/drug effects , Nuclear Matrix/metabolism , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Proteins
11.
Nature ; 408(6814): 873-6, 2000 Dec 14.
Article in English | MEDLINE | ID: mdl-11130728

ABSTRACT

The considerable length of DNA in eukaryotic genomes requires packaging into chromatin to fit inside the small dimensions of the cell nucleus. Histone H1 functions in the compaction of chromatin into higher order structures derived from the repeating 'beads on a string' nucleosome polymer. Modulation of H1 binding activity is thought to be an important step in the potentiation/depotentiation of chromatin structure for transcription. It is generally accepted that H1 binds less tightly than other histones to DNA in chromatin and can readily exchange in living cells. Fusion proteins of Histone H1 and green fluorescent protein (GFP) have been shown to associate with chromatin in an apparently identical fashion to native histone H1. This provides a means by which to study histone H1-chromatin interactions in living cells. Here we have used human cells with a stably integrated H1.1-GFP fusion protein to monitor histone H1 movement directly by fluorescence recovery after photobleaching in living cells. We find that exchange is rapid in both condensed and decondensed chromatin, occurs throughout the cell cycle, and does not require fibre-fibre interactions. Treatment with drugs that alter protein phosphorylation significantly reduces exchange rates. Our results show that histone H1 exchange in vivo is rapid, occurs through a soluble intermediate, and is modulated by the phosphorylation of a protein or proteins as yet to be determined.


Subject(s)
Chromatin/metabolism , Histones/metabolism , DNA/metabolism , Dichlororibofuranosylbenzimidazole/pharmacology , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins , Humans , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Phosphorylation , Protein Binding , Protein Kinase Inhibitors , Protein Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Staurosporine/pharmacology , Tumor Cells, Cultured
12.
J Cell Biol ; 150(1): 41-51, 2000 Jul 10.
Article in English | MEDLINE | ID: mdl-10893255

ABSTRACT

Compartmentalization of the nucleus is now recognized as an important level of regulation influencing specific nuclear processes. The mechanism of factor organization and the movement of factors in nuclear space have not been fully determined. Splicing factors, for example, have been shown to move in a directed manner as large intact structures from sites of concentration to sites of active transcription, but splicing factors are also thought to exist in a freely diffusible state. In this study, we examined the movement of a splicing factor, ASF, green fluorescent fusion protein (ASF-GFP) using time-lapse microscopy and the technique fluorescence recovery after photobleaching (FRAP). We find that ASF-GFP moves at rates up to 100 times slower than free diffusion when it is associated with speckles and, surprisingly, also when it is dispersed in the nucleoplasm. The mobility of ASF is consistent with frequent but transient interactions with relatively immobile nuclear binding sites. This mobility is slightly increased in the presence of an RNA polymerase II transcription inhibitor and the ASF molecules further enrich in speckles. We propose that the nonrandom organization of splicing factors reflects spatial differences in the concentration of relatively immobile binding sites.


Subject(s)
Cell Compartmentation/physiology , Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Animals , Binding Sites , Biological Transport/drug effects , Biological Transport/physiology , Cell Line , Cell Nucleus/ultrastructure , Diffusion , Enzyme Inhibitors/pharmacology , Fluorescence , Genes, Reporter , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mice , Muntjacs , Photochemistry , Protein Kinase Inhibitors , RNA Polymerase II/antagonists & inhibitors , RNA-Binding Proteins , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine-Arginine Splicing Factors , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/drug effects
13.
J Cell Biol ; 148(2): 283-92, 2000 Jan 24.
Article in English | MEDLINE | ID: mdl-10648561

ABSTRACT

The promyelocytic leukemia (PML) nuclear body (also referred to as ND10, POD, and Kr body) is involved in oncogenesis and viral infection. This subnuclear domain has been reported to be rich in RNA and a site of nascent RNA synthesis, implicating its direct involvement in the regulation of gene expression. We used an analytical transmission electron microscopic method to determine the structure and composition of PML nuclear bodies and the surrounding nucleoplasm. Electron spectroscopic imaging (ESI) demonstrates that the core of the PML nuclear body is a dense, protein-based structure, 250 nm in diameter, which does not contain detectable nucleic acid. Although PML nuclear bodies contain neither chromatin nor nascent RNA, newly synthesized RNA is associated with the periphery of the PML nuclear body, and is found within the chromatin-depleted region of the nucleoplasm immediately surrounding the core of the PML nuclear body. We further show that the RNA does not accumulate in the protein core of the structure. Our results dismiss the hypothesis that the PML nuclear body is a site of transcription, but support the model in which the PML nuclear body may contribute to the formation of a favorable nuclear environment for the expression of specific genes.


Subject(s)
Cell Nucleus/ultrastructure , Leukemia, Promyelocytic, Acute/pathology , Neoplasm Proteins/isolation & purification , RNA, Neoplasm/isolation & purification , RNA, Nuclear/isolation & purification , Acetylation , Chromatin/chemistry , Chromatin/ultrastructure , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Microscopy, Electron/methods , Microtomy , Nitrogen/isolation & purification , Organometallic Compounds , Phosphorus/isolation & purification , Spectrum Analysis , Staining and Labeling/methods
14.
Proc Natl Acad Sci U S A ; 96(21): 11883-8, 1999 Oct 12.
Article in English | MEDLINE | ID: mdl-10518545

ABSTRACT

In analyzing the transcriptional networks that regulate development, one ideally would like to determine whether a particular transcription factor binds directly to a candidate target promoter inside the living embryo. Properties of the Caenorhabditis elegans elt-2 gene, which encodes a gut-specific GATA factor, have allowed us to develop such a method. We previously have shown, by means of ectopic expression studies, that elt-2 regulates its own promoter. To test whether this autoregulation is direct, we fused green fluorescent protein (GFP) close to the C terminus of elt-2 in a construct that contains the full elt-2 promoter and the full elt-2 zinc finger DNA binding domain; the construct is expressed correctly (i.e., only in the gut lineage) and is able to rescue the lethality of an elt-2 null mutant. Multicopy transgenic arrays of this rescuing elt-2::GFP construct were integrated into the genome and transgenic embryos were examined when the developing gut has 4-8 cells; the majority of these embryonic gut nuclei show two discrete intense foci of fluorescence. We interpret these fluorescent foci as the result of ELT-2::GFP binding directly to its own promoter within nuclei of the developing gut lineage. Numerous control experiments, both genetic and biochemical, all support this conclusion and support the specificity of the binding. The approach should be applicable to studying other transcription factors binding target promoters, all within the living C. elegans embryo.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans/embryology , Microscopy, Video/methods , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Cell Nucleus/metabolism , Female , GATA Transcription Factors , Green Fluorescent Proteins , Intestinal Mucosa/metabolism , Intestines/embryology , Luminescent Proteins/metabolism , Male , Models, Genetic , Plasmids/metabolism , Promoter Regions, Genetic , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription Factors/genetics
15.
J Biol Chem ; 274(35): 24914-20, 1999 Aug 27.
Article in English | MEDLINE | ID: mdl-10455166

ABSTRACT

When the Ras mitogen-activated protein kinase (MAPK) signaling pathway of quiescent cells is stimulated with growth factors or phorbol esters, the early response genes c-fos and c-myc are rapidly induced, and concurrently there is a rapid phosphorylation of histone H3. Using an antibody specific for phosphorylated Ser-10 of H3, we show that Ser-10 of H3 is phosphorylated, and we provide direct evidence that phosphorylated H3 is associated with c-fos and c-myc genes in stimulated cells. H3 phosphorylation may contribute to proto-oncogene induction by modulating chromatin structure and releasing blocks in elongation. Previously we reported that persistent stimulation of the Ras-MAPK signaling pathway in oncogene-transformed cells resulted in increased amounts of phosphorylated histone H1. Here we show that phosphorylated H3 is elevated in the oncogene-transformed mouse fibroblasts. Further we show that induction of ras expression results in a rapid increase in H3 phosphorylation. H3 phosphatase, identified as PP1, activities in ras-transformed and parental fibroblast cells were similar, suggesting that elevated H3 kinase activity was responsible for the increased level of phosphorylated H3 in the oncogene-transformed cells. Elevated levels of phosphorylated H1 and H3 may be responsible for the less condensed chromatin structure and aberrant gene expression observed in the oncogene-transformed cells.


Subject(s)
Histones/metabolism , Mitogens/pharmacology , Phosphoserine/metabolism , Proto-Oncogene Proteins/genetics , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Chromatin/metabolism , Epidermal Growth Factor/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation , Genes, fos , Genes, myc , Genes, ras , Mice , Okadaic Acid/pharmacology , Phosphorylation , Phosphoserine/immunology , Tetradecanoylphorbol Acetate/pharmacology , Transformation, Genetic
16.
Micron ; 30(2): 151-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10420641

ABSTRACT

We describe a method to image selectively the protein-based architecture in the eukaryotic cell nucleus using nitrogen and phosphorus mapping. In addition, we describe a method to determine total mass as well as stoichiometric relationships between protein and RNA. This method is illustrated using particulate structures in the nucleus called interchromatin granules. In so doing, we demonstrate that these granules contain heterogeneous nuclear RNA, and have an average protein and RNA content of 3.094 and 1.672 MDa, respectively. We also tested the sensitivity of phosphorus detection by exogenously applying purified duplex DNA to the surfaces of thin sections, and have shown that structures as small as single molecules of duplex DNA can be detected in situ using these electron spectroscopic imaging techniques.


Subject(s)
Cell Nucleus/chemistry , Nuclear Proteins/analysis , RNA, Nuclear/analysis , Animals , Cell Line , Cell Nucleus/ultrastructure , Chromatin/metabolism , Chromatin/ultrastructure , Fibroblasts , Microscopy, Electron/methods , Muntjacs , Nitrogen/metabolism , Phosphorus/metabolism , Ribosomes/metabolism
17.
Mol Biol Cell ; 10(6): 2051-62, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10359614

ABSTRACT

Whether the cell nucleus is organized by an underlying architecture analagous to the cytoskeleton has been a highly contentious issue since the original isolation of a nuclease and salt-resistant nuclear matrix. Despite electron microscopy studies that show that a nuclear architecture can be visualized after fractionation, the necessity to elute chromatin to visualize this structure has hindered general acceptance of a karyoskeleton. Using an analytical electron microscopy method capable of quantitative elemental analysis, electron spectroscopic imaging, we show that the majority of the fine structure within interchromatin regions of the cell nucleus in fixed whole cells is not nucleoprotein. Rather, this fine structure is compositionally similar to known protein-based cellular structures of the cytoplasm. This study is the first demonstration of a protein network in unfractionated and uninfected cells and provides a method for the ultrastructural characterization of the interaction of this protein architecture with chromatin and ribonucleoprotein elements of the cell nucleus.


Subject(s)
Cell Nucleus/ultrastructure , Microscopy, Electron/methods , Nuclear Proteins/metabolism , Nuclear Proteins/ultrastructure , Animals , Cell Nucleus/chemistry , Chromatin/ultrastructure , Fixatives/chemistry , Formaldehyde/chemistry , Image Enhancement , Molecular Biology/methods , Nitrogen , Nucleic Acids/metabolism , Nucleic Acids/ultrastructure , Phosphorus , Phosphorylation , Polymers/chemistry , Reproducibility of Results
18.
J Biol Chem ; 274(17): 11713-20, 1999 Apr 23.
Article in English | MEDLINE | ID: mdl-10206986

ABSTRACT

Histone deacetylases are the catalytic subunits of multiprotein complexes that are targeted to specific promoters through their interaction with sequence-specific DNA-binding factors. We have cloned and characterized a new human cDNA, HDAC-A, with homology to the yeast HDA1 family of histone deacetylases. Analysis of the predicted amino acid sequence of HDAC-A revealed an open reading frame of 967 amino acids containing two domains: a NH2-terminal domain with no homology to known proteins and a COOH-terminal domain with homology to known histone deacetylases (42% similarity to RPD3, 60% similarity to HDA1). Three additional human cDNAs with high homology to HDAC-A were identified in sequence data bases, indicating that HDAC-A itself is a member of a new family of human histone deacetylases. The mRNA encoding HDAC-A was differentially expressed in a variety of human tissues. The expressed protein, HDAC-Ap, exhibited histone deacetylase activity and this activity mapped to the COOH-terminal region (amino acids 495-967) with homology to HDA1p. In immunoprecipitation experiments, HDAC-A interacted specifically with several cellular proteins, indicating that it might be part of a larger multiprotein complex.


Subject(s)
Histone Deacetylases/genetics , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary , Humans , Molecular Sequence Data , RNA, Messenger/genetics , Sequence Homology, Amino Acid
19.
Methods ; 17(2): 188-200, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10075896

ABSTRACT

The analytical electron microscope technique called electron spectroscopic imaging (ESI) has a number of applications in the study of DNA:protein complexes. The method offers an intermediate level of spatial resolution for in vitro structural studies of complexes that may be too large or heterogeneous to study by crystallography or magnetic resonance spectroscopy. An advantage of ESI is that the distribution of nucleic acids can be resolved in a nucleoprotein complex by mapping the element phosphorus, present at high levels in nucleic acid compared to protein. Measurements of phosphorus content together with mass determination allows estimates to be made of stoichiometric relationships of protein and nucleic acids in these complexes. ESI is also suited to in situ studies of nuclear structure. Mass-sensitive images combined with nitrogen and phosphorus maps can be used to distinguish nucleic acid components from nuclear structures that are predominantly protein based. Interactions between chromatin on the periphery of interchromatin granule clusters (IGC) with the protein substructure that connects the exterior of the IGC to its core can be studied with this technique. The method also avoids the use of heavy atom stains, agents required in conventional electron microscopy, that preclude the distinguishing of structures on the basis of their biochemical composition. The principles of ESI and technical aspects of the method are discussed.


Subject(s)
Chromatin/ultrastructure , Microscopy, Electron/methods , Animals , Chromatin/metabolism , DNA/analysis , Fibroblasts/metabolism , Fibroblasts/ultrastructure , HeLa Cells , Humans , Nitrogen/metabolism , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Phosphorus/metabolism , Proteins/analysis , Xenopus/metabolism
20.
J Biol Chem ; 273(38): 24470-8, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733739

ABSTRACT

Apoptosis plays an important role in the survival of an organism, and substantial work has been done to understand the signaling pathways that regulate this process. Characteristic changes in chromatin organization accompany apoptosis and are routinely used as markers for cell death. We have examined the organization of chromatin in apoptotic PC12 and HeLa cells by indirect immunofluorescence and electron spectroscopic imaging. Our results indicate that de novo chromatin condensation normally seen during mitosis does not occur when cells undergo apoptosis. Instead, the condensed chromatin typically observed results from aggregation of the heterochromatin. We present evidence that, early in apoptosis, there is a rapid degradation of the nuclease-hypersensitive euchromatin that contains hyperacetylated histones. This occurs coincident with the loss of nuclear integrity due to degradation of lamins and reorganization of intranuclear protein matrix. These events lead to collapse of the nucleus and aggregation of heterochromatin to produce the appearance of condensed apoptotic chromatin. This heterochromatin aggregate is then digested by nucleases to produce the oligonucleosomal DNA ladder that is a hallmark of late apoptosis. Unlike mitosis, we have not seen any evidence for the requirement of phosphorylated histones H1 and H3 to maintain the chromatin in the condensed state.


Subject(s)
Apoptosis/physiology , Chromatin/physiology , Heterochromatin/physiology , Animals , Apoptosis/drug effects , Butyrates/pharmacology , Butyric Acid , Cell Survival , Chromatin/ultrastructure , DNA/metabolism , Euchromatin , Fluorescent Antibody Technique, Indirect , HeLa Cells , Heterochromatin/ultrastructure , Histones/metabolism , Humans , Mitosis , Models, Biological , Nucleosomes/physiology , Nucleosomes/ultrastructure , PC12 Cells , Phosphorylation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL