Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 19(6): 1089-97, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11713576

ABSTRACT

Cells from individuals with Cockayne syndrome (CS) have a defect in transcription-coupled DNA repair (TCR), which rapidly corrects certain DNA lesions located on the transcribed strand of active genes. Despite this DNA repair defect, individuals with CS (of which there are two complementation groups, CSA and CSB) do not demonstrate an elevated incidence of cancer. Recently, we demonstrated that disruption of the CSB gene reduces the spontaneous tumor rate in cancer predisposed Ink4a/ARF-/- mice as well as causing their embryo fibroblasts to proliferate more slowly and be more sensitive to UV-induced apoptosis. In the present study we characterized phosphorothioate backbone antisense oligodeoxynucleotides (AOs) that reduced the levels of CSB mRNA in A2780/CP70 ovarian carcinoma cells. The AOs caused the cells to proliferate more slowly and made them more sensitive to either cisplatin or oxaliplatin. The AOs also enhanced the cytotoxicity of hydrogen peroxide and gamma-radiation, both of which can induce oxidative DNA lesions, which are subject to TCR. The AOs did not potentiate the cytotoxicity of topotecan, which induces DNA strand breaks. Chemically modified () AOs (MBOs) targeting CSB were able to potentiate the anti-tumor effect of cisplatin against A2780/CP70 tumor xenografts formed in nude mice. The MBOs enabled a non-toxic (3 mg/kg) dose of cisplatin to have the same degree of anti-tumor efficacy as a more toxic (5 mg/kg) cisplatin dose. Collectively, these results suggest that the CSB gene product may be viewed as an anti-cancer target.


Subject(s)
DNA Helicases/genetics , DNA Repair , Oligonucleotides, Antisense/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cisplatin/pharmacology , Cockayne Syndrome/genetics , DNA Helicases/metabolism , DNA Primers/chemistry , DNA Repair Enzymes , Drug Resistance , Drug Synergism , Female , Fibroblasts/metabolism , Gene Expression , Humans , Mice , Mice, Nude , Organoplatinum Compounds/pharmacology , Ovarian Neoplasms/pathology , Oxaliplatin , Poly-ADP-Ribose Binding Proteins , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thionucleotides/therapeutic use , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects
2.
Mutat Res ; 486(4): 259-74, 2001 Sep 04.
Article in English | MEDLINE | ID: mdl-11516929

ABSTRACT

It has been shown that ultraviolet (UV) radiation induces the ubiquitination of the large subunit of RNA polymerase II (RNAP II-LS) as well as its proteasomal degradation. Studies in mammalian cells have indicated that highly phosphorylated forms of RNAP II-LS are preferentially ubiquitinated, but studies in Saccharomyces cerevisiae have provided evidence that unphosphorylated RNAP II-LS is an equally suitable substrate. In the present study, an antibody (ARNA-3) that recognizes all forms of RNAP II-LS, regardless of the phosphorylation status of its C-terminal domain (CTD), was utilized to evaluate the degradation of total cellular RNAP II-LS in human fibroblasts under basal conditions or after UV-C (10J/m(2)) irradiation. It was found that UV radiation rapidly shifted the phosphorylation profile of RNAP II-LS from a mixture of dephosphorylated and phosphorylated forms to entirely more phosphorylated forms. This shift in phosphorylation status was not blocked by pharmacologic inhibition of either the ERK or p38 pathways, both of which have been implicated in the cellular UV response. In addition to shifting the phosphorylation profile, UV radiation led to net degradation of total RNAP II-LS. UV-induced degradation of RNAP II-LS was also greatly reduced in the presence of the transcriptional and CTD kinase inhibitor DRB. Using a panel of protease inhibitors, it was shown that the bulk of UV-induced degradation is proteasome-dependent. However, the UV-induced loss of hypophosphorylated RNAP II-LS was proteasome-independent. Lastly, UV radiation induced a similar shift to all hyperphosphorylated RNAP II-LS in Cockayne syndrome (CS) cells of complementation groups A or B (CSA or CSB) when compared to appropriate controls. The UV-induced degradation rates of RNAP II-LS were not significantly altered when comparing CSA or CSB to repair competent control cells. The implications for the cellular UV response are discussed.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Ultraviolet Rays , Cell Line , Dichlororibofuranosylbenzimidazole/pharmacology , Down-Regulation , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Immunoblotting , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/radiation effects , Proteasome Endopeptidase Complex , Protein Structure, Tertiary , Time Factors , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases
3.
Mol Cell Biol ; 21(5): 1810-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238917

ABSTRACT

Cells isolated from individuals with Cockayne syndrome (CS) have a defect in transcription-coupled DNA repair, which rapidly corrects certain DNA lesions located on the transcribed strand of active genes. Despite this DNA repair defect, individuals with CS group A (CSA) or group B (CSB) do not exhibit an increased spontaneous or UV-induced cancer rate. In order to investigate the effect of CSB deficiency on spontaneous carcinogenesis, we crossed CSB(-/-) mice with cancer-prone mice lacking the p16(Ink4a)/p19(ARF) tumor suppressor locus. CSB(-/-) mice are sensitive to UV-induced skin cancer but show no increased rate of spontaneous cancer. CSB(-/-) Ink4a/ARF(-/-) mice developed 60% fewer tumors than Ink4a/ARF(-/-) animals and demonstrated a longer tumor-free latency time (260 versus 150 days). Moreover, CSB(-/-) Ink4a/ARF(-/-) mouse embryo fibroblasts (MEFs) exhibited a lower colony formation rate after low-density seeding, a lower rate of H-Ras-induced transformation, slower proliferation, and a lower mRNA synthesis rate than Ink4a/ARF(-/-) MEFs. CSB(-/-) Ink4a/ARF(-/-) MEFs were also more sensitive to UV-induced p53 induction and UV-induced apoptosis than were Ink4a/ARF(-/-) MEFs. In order to investigate whether the apparent antineoplastic effect of CSB gene disruption was caused by sensitization to genotoxin-induced (p53-mediated) apoptosis or by p53-independent sequelae, we also generated p53(-/-) and CSB(-/-) p53(-/-) MEFs. The CSB(-/-) p53(-/-) MEFs demonstrated lower colony formation efficiency, a lower proliferation rate, a lower mRNA synthesis rate, and a higher rate of UV-induced cell death than p53(-/-) MEFs. Collectively, these results indicate that the antineoplastic effect of CSB gene disruption is at least partially p53 independent; it may result from impaired transcription or from apoptosis secondary to environmental or endogenous DNA damage.


Subject(s)
Cockayne Syndrome/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Helicases/genetics , DNA Helicases/physiology , Neoplasms/genetics , Proteins/genetics , Age Factors , Animals , Apoptosis , Cell Division , Crosses, Genetic , DNA Repair , DNA Repair Enzymes , Fibroblasts/metabolism , Fibrosarcoma/metabolism , Flow Cytometry , Genes, p53/genetics , Genetic Predisposition to Disease , Genotype , Immunoblotting , In Situ Nick-End Labeling , Lymphoma/metabolism , Mice , Mice, Knockout , Poly-ADP-Ribose Binding Proteins , RNA, Messenger/metabolism , Time Factors , Transformation, Genetic , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , ras Proteins/metabolism
4.
J Neuropathol Exp Neurol ; 60(12): 1219-32, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11764094

ABSTRACT

Affected neurons of Alzheimer disease (AD) brain are distinguished by the presence of the cell cycle cdc2 kinase and mitotic phosphoepitopes. A significant body of previous data has documented a decrease in neuronal RNA levels and nucleolar volume in AD brain. Here we present evidence that integrates these seemingly distinct findings and offers an explanation for the degenerative outcome of the disease. During mitosis cdc2 phosphorylates and inhibits the major transcriptional regulator RNA polymerase II (RNAP II). We therefore investigated cdc2 phosphorylation of RNAP II in AD brain. Using the H5 and H14 monoclonal antibodies specific for the cdc2-phosphorylated sites in RNAP II, we found that the polymerase is highly phosphorylated in AD. Moreover, RNAP II in AD translocates from its normally nuclear compartment to the cytoplasm of affected neurons, where it colocalizes with cdc2. These M phase-like changes in RNAP II correlate with decreased levels of poly-A RNA in affected neurons. Significantly, they precede tau phosphorylation and neurofibrillary tangle formation. Our data support the hypothesis that inappropriate activation of the cell cycle cdc2 kinase in differentiated neurons contributes to neuronal dysfunction and degeneration in part by inhibiting RNAP II and cellular processes dependent on transcription.


Subject(s)
Alzheimer Disease/enzymology , Neurofibrillary Tangles/enzymology , Neurons/enzymology , RNA Polymerase II/metabolism , RNA/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/enzymology , Brain/metabolism , Brain/pathology , Female , Humans , Male , Mice , Middle Aged , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Neurons/metabolism , Neurons/pathology , Phosphorylation
5.
Mol Endocrinol ; 14(11): 1750-75, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11075810

ABSTRACT

Caveolin-1 was first identified as a phosphoprotein in Rous sarcoma virus (RSV)-transformed chicken embryo fibroblasts. Tyrosine 14 is now thought to be the principal site for recognition by c-Src kinase; however, little is known about this phosphorylation event. Here, we generated a monoclonal antibody (mAb) probe that recognizes only tyrosine 14-phosphorylated caveolin-1. Using this approach, we show that caveolin-1 (Y14) is a specific tyrosine kinase substrate that is constitutively phosphorylated in Src- and Abl-transformed cells and transiently phosphorylated in a regulated fashion during growth factor signaling. We also provide evidence that tyrosine-phosphorylated caveolin-1 is localized at the major sites of tyrosine-kinase signaling, i.e. focal adhesions. By analogy with other signaling events, we hypothesized that caveolin-1 could serve as a docking site for pTyr-binding molecules. In support of this hypothesis, we show that phosphorylation of caveolin-1 on tyrosine 14 confers binding to Grb7 (an SH2-domain containing protein) both in vitro and in vivo. Furthermore, we demonstrate that binding of Grb7 to tyrosine 14-phosphorylated caveolin-1 functionally augments anchorage-independent growth and epidermal growth factor (EGF)-stimulated cell migration. We discuss the possible implications of our findings in the context of signal transduction.


Subject(s)
Caveolins/metabolism , Growth Substances/metabolism , Proteins/metabolism , Tyrosine/metabolism , src-Family Kinases/metabolism , 3T3 Cells , Adipocytes/drug effects , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Caveolae/metabolism , Caveolin 1 , Caveolins/genetics , Caveolins/immunology , Cell Adhesion/physiology , Cell Division/physiology , Cell Movement/physiology , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Female , GRB7 Adaptor Protein , Humans , Insulin/metabolism , Insulin/pharmacology , Lipid Metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phosphorylation/drug effects , Signal Transduction , Vanadates/pharmacology
6.
Proc Natl Acad Sci U S A ; 97(19): 10503-8, 2000 Sep 12.
Article in English | MEDLINE | ID: mdl-10973477

ABSTRACT

Cells from patients with Cockayne syndrome (CS) are hypersensitive to DNA-damaging agents and are unable to restore damage-inhibited RNA synthesis. On the basis of repair kinetics of different types of lesions in transcriptionally active genes, we hypothesized previously that impaired transcription in CS cells is a consequence of defective transcription initiation after DNA damage induction. Here, we investigated the effect of UV irradiation on transcription by using an in vitro transcription system that allowed uncoupling of initiation from elongation events. Nuclear extracts prepared from UV-irradiated or mock-treated normal human and CS cells were assayed for transcription activity on an undamaged beta-globin template. Transcription activity in nuclear extracts closely mimicked kinetics of transcription in intact cells: extracts from normal cells prepared 1 h after UV exposure showed a strongly reduced activity, whereas transcription activity was fully restored in extracts prepared 6 h after treatment. Extracts from CS cells exhibited reduced transcription activity at any time after UV exposure. Reduced transcription activity in extracts coincided with a strong reduction of RNA polymerase II (RNAPII) containing hypophosphorylated C-terminal domain, the form of RNAPII known to be recruited to the initiation complex. These results suggest that inhibition of transcription after UV irradiation is at least partially caused by repression of transcription initiation and not solely by blocked elongation at sites of lesions. Generation of hypophosphorylated RNAPII after DNA damage appears to play a crucial role in restoration of transcription. CS proteins may be required for this process in a yet unknown way.


Subject(s)
RNA Polymerase II/metabolism , Transcription, Genetic/radiation effects , Ultraviolet Rays , Base Sequence , Blotting, Western , Cell Extracts , Cell Line, Transformed , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Primers , Genetic Complementation Test , Humans , Phosphorylation
7.
J Biol Chem ; 275(48): 37702-11, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-10973975

ABSTRACT

Caveolin-3 is the principal structural protein of caveolae in striated muscle. Autosomal dominant limb-girdle muscular dystrophy (LGMD-1C) in humans is due to mutations (DeltaTFT and Pro --> Leu) within the CAV3 gene. We have shown that LGMD-1C mutations lead to formation of unstable aggregates of caveolin-3 that are retained intracellularly and are rapidly degraded. The mechanism by which LGMD-1C mutants of caveolin-3 are degraded remains unknown. Here, we show that LGMD-1C mutants of caveolin-3 undergo ubiquitination-proteasomal degradation. Treatment with proteasomal inhibitors (MG-132, MG-115, lactacystin, or proteasome inhibitor I), but not lysosomal inhibitors, prevented degradation of LGMD-1C caveolin-3 mutants. In the presence of MG-132, LGMD-1C caveolin-3 mutants accumulated within the endoplasmic reticulum and did not reach the plasma membrane. LGMD-1C mutants of caveolin-3 behave in a dominant negative fashion, causing intracellular retention and degradation of wild-type caveolin-3. Interestingly, in cells co-expressing wild-type and mutant forms of caveolin-3, MG-132 treatment rescued wild-type caveolin-3; wild-type caveolin-3 was not degraded and reached the plasma membrane. These results may have clinical implications for treatment of patients with LGMD-1C.


Subject(s)
Caveolins/metabolism , Cysteine Endopeptidases/metabolism , Cytoskeletal Proteins/metabolism , Membrane Glycoproteins/metabolism , Multienzyme Complexes/metabolism , Muscular Dystrophies/genetics , Mutation , Ubiquitins/metabolism , 3T3 Cells , Animals , Caveolin 3 , Caveolins/genetics , Cysteine Proteinase Inhibitors/pharmacology , Cytoskeletal Proteins/chemistry , Hydrolysis , Membrane Glycoproteins/chemistry , Mice , Multienzyme Complexes/antagonists & inhibitors , Proteasome Endopeptidase Complex
8.
Front Biosci ; 5: D244-57, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10704151

ABSTRACT

The cell cycle and transcription by RNA polymerase II (RNAP II) are closely related. They utilize shared components. RNAP II transcriptional activity is modulated during the cell cycle. Cell cycle dependent changes in the phosphorylation status of the carboxyl-terminal domain (CTD) of the largest subunit of RNAP II (RNAP II-LS) alter transcription. Several CTD kinases are members of the cyclin-dependent kinase (cdk) superfamily, including p34cdc2 (cdk1), cdk7, cdk8, and cdk9. Each of these cdks, with their respective cyclin partners, have been linked to cell cycle regulatory events. Other CTD kinases such as casein kinase II (CKII) and c-abl have also been implicated in cell cycle dependent modifications of the CTD. In addition, the stalling of RNAP II complexes at DNA lesions helps stimulate p53 accumulation which largely determines the cell's DNA damage response, including cell cycle arrest. Alzheimer's disease pathology results partially from activation of mitotic cdks in postmitotic neurons which can phosphorylate RNAP II-LS and other targets.


Subject(s)
Cell Cycle/physiology , Cyclin-Dependent Kinases/physiology , RNA Polymerase II/physiology , Animals , CDC2 Protein Kinase/physiology , Casein Kinase II , DNA Polymerase II/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Kinases/metabolism , Protein Kinases/physiology , Protein Serine-Threonine Kinases/metabolism , Transcription Factor TFIID , Transcription Factors, TFII/metabolism
9.
J Biol Chem ; 274(39): 27823-8, 1999 Sep 24.
Article in English | MEDLINE | ID: mdl-10488128

ABSTRACT

Monoclonal antibodies that recognize specific carboxyl-terminal domain (CTD) phosphoepitopes were used to examine CTD phosphorylation in yeast cells lacking carboxyl-terminal domain kinase I (CTDK-I). We show that deletion of the kinase subunit CTK1 results in an increase in phosphorylation of serine in position 5 (Ser(5)) of the CTD repeat (Tyr(1)-Ser(2)-Pro(3)-Thr(4)-Ser(5)-Pro(6)-Ser(7)) during logarithmic growth. This result indicates that CTDK-I negatively regulates CTD Ser(5) phosphorylation. We also show that CTK1 deletion (ctk1Delta) eliminates the transient increase in CTD serine 2 (Ser(2)) phosphorylation observed during the diauxic shift. This result suggests that CTDK-I may play a direct role in phosphorylating CTD Ser(2) in response to nutrient depletion. Northern blot analysis was used to show that genes normally induced during the diauxic shift are not properly induced in a ctk1Delta strain. Glycogen synthase (GSY2) and cytosolic catalase (CTT1) mRNA levels increase about 10-fold in wild-type cells, but this increase is not observed in ctk1Delta cells suggesting that increased message levels may require Ser(2) phosphorylation. Heat shock also induces Ser(2) phosphorylation, but we show here that this change in CTD modification and an accompanying induction of heat shock gene expression is independent of CTDK-I. The observation that SSA3/SSA4 expression is increased in ctk1Delta cells grown at normal temperature suggests a possible role for CTDK-I in transcription repression. We discuss several possible positive and negative roles for CTDK-I in regulating CTD phosphorylation and gene expression.


Subject(s)
Gene Expression Regulation, Fungal , Protein Kinases/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/enzymology , Catalase/genetics , Cloning, Molecular , Cytosol/enzymology , Gene Expression Regulation, Enzymologic , Glycogen Synthase/genetics , Hot Temperature , Kinetics , Macromolecular Substances , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Phosphorylation , Protein Kinases/genetics , RNA Polymerase II/chemistry , RNA, Messenger/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Sequence Deletion , Transcription, Genetic
10.
J Biol Chem ; 273(9): 5184-9, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9478972

ABSTRACT

We have shown previously that UV radiation and other DNA-damaging agents induce the ubiquitination of a portion of the RNA polymerase II large subunit (Pol II LS). In the present study UV irradiation of repair-competent fibroblasts induced a transient reduction of the Pol II LS level; new protein synthesis restored Pol II LS to the base-line level within 16-24 h. In repair-deficient xeroderma pigmentosum cells, UV radiation-induced ubiquitination of Pol II LS was followed by a sustained reduction of Pol II LS level. In both normal and xeroderma pigmentosum cells, the ubiquitinated Pol II LS had a hyperphosphorylated COOH-terminal domain (CTD), which is characteristic of elongating Pol II. The portion of Pol II LS whose steady-state level diminished most quickly had a relatively hypophosphorylated CTD. The ubiquitinated residues did not map to the CTD. Importantly, UV-induced reduction of Pol II LS level in repair-competent or -deficient cells was inhibited by the proteasome inhibitors lactacystin or MG132. These data demonstrate that UV-induced ubiquitination of Pol II LS is followed by its degradation in the proteasome. These results suggest, contrary to a current model of transcription-coupled DNA repair, that elongating Pol II complexes which arrest at intragenic DNA lesions may be aborted rather than resuming elongation after repair takes place.


Subject(s)
Cysteine Endopeptidases/metabolism , DNA Repair , Multienzyme Complexes/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic/radiation effects , Ubiquitins/metabolism , Cell Line , Cysteine Proteinase Inhibitors , Humans , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational/radiation effects , Ultraviolet Rays , Xeroderma Pigmentosum/metabolism
11.
J Cell Biol ; 136(1): 19-28, 1997 Jan 13.
Article in English | MEDLINE | ID: mdl-9008700

ABSTRACT

The carboxy-terminal domain (CTD) of the largest subunit of RNA polymerase II (Pol II) contains multiple tandem copies of the consensus heptapeptide, TyrSerProThrSerProSer. Concomitant with transcription initiation the CTD is phosphorylated. Elongating polymerase has a hyperphosphorylated CTD, but the role of this modification is poorly understood. A recent study revealed that some hyperphosphorylated polymerase molecules (Pol IIo) are nonchromosomal, and hence transcriptionally unengaged (Bregman, D.B., L. Du, S. van der Zee, S.L. Warren. 1995. J. Cell Biol. 129: 287-298). Pol IIo was concentrated in discrete splicing factor domains, suggesting a possible relationship between CTD phosphorylation and splicing factors, but no evidence beyond immunolocalization data was provided to support this idea. Here, we show that Pol IIo co-immunoprecipitates with members of two classes of splicing factors, the Sm snRNPs and non-snRNP SerArg (SR) family proteins. Significantly, Pol IIo's association with splicing factors is maintained in the absence of pre-mRNA, and the polymerase need not be transcriptionally engaged. We also provide definitive evidence that hyperphosphorylation of Pol II's CTD is poorly correlated with its transcriptional activity. Using monoclonal antibodies (mAbs) H5 and H14, which are shown here to recognize phosphoepitopes on Pol II's CTD, we have quantitated the level of Pol IIo at different stages of the cell cycle. The level of Pol IIo is similar in interphase and mitotic cells, which are transcriptionally active and inactive, respectively. Finally, complexes containing Pol IIo and splicing factors can be prepared from mitotic as well as interphase cells. The experiments reported here establish that hyperphosphorylation of the CTD is a good indicator of polymerase's association with snRNP and SR splicing factors, but not of its transcriptional activity. Most importantly, the present study suggests that splicing factors may associate with the polymerase via the hyperphosphorylated CTD.


Subject(s)
RNA Polymerase II/metabolism , RNA Splicing/physiology , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Cell Cycle , HeLa Cells , Humans , Mitosis , Phosphorylation , Precipitin Tests , RNA Polymerase II/genetics , RNA Precursors/analysis , Recombinant Fusion Proteins , Transcription, Genetic/physiology
12.
Proc Natl Acad Sci U S A ; 93(21): 11586-90, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8876179

ABSTRACT

Damage to actively transcribed DNA is preferentially repaired by the transcription-coupled repair (TCR) system. TCR requires RNA polymerase II (Pol II), but the mechanism by which repair enzymes preferentially recognize and repair DNA lesions on Pol II-transcribed genes is incompletely understood. Herein we demonstrate that a fraction of the large subunit of Pol II (Pol II LS) is ubiquitinated after exposing cells to UV-radiation or cisplatin but not several other DNA damaging agents. This novel covalent modification of Pol II LS occurs within 15 min of exposing cells to UV-radiation and persists for about 8-12 hr. Ubiquitinated Pol II LS is also phosphorylated on the C-terminal domain. UV-induced ubiquitination of Pol II LS is deficient in fibroblasts from individuals with two forms of Cockayne syndrome (CS-A and CS-B), a rare disorder in which TCR is disrupted. UV-induced ubiquitination of Pol II LS can be restored by introducing cDNA constructs encoding the CSA or CSB genes, respectively, into CS-A or CS-B fibroblasts. These results suggest that ubiquitination of Pol II LS plays a role in the recognition and/or repair of damage to actively transcribed genes. Alternatively, these findings may reflect a role played by the CSA and CSB gene products in transcription.


Subject(s)
Cockayne Syndrome/enzymology , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Ubiquitins/metabolism , Ultraviolet Rays , Cell Line , Cisplatin/pharmacology , Cockayne Syndrome/genetics , DNA Damage , DNA Repair , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Phosphorylation , RNA Polymerase II/radiation effects , Recombinant Proteins/metabolism , Recombinant Proteins/radiation effects , Transcription, Genetic , Transfection
13.
J Cell Biol ; 129(2): 287-98, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7536746

ABSTRACT

A subpopulation of the largest subunit of RNA polymerase II (Pol II LS) is located in 20-50 discrete subnuclear domains that are closely linked to speckle domains, which store splicing proteins. The speckle-associated fraction of Pol II LS is hyperphosphorylated on the COOH-terminal domain (CTD), and it is highly resistant to extraction by detergents. A diffuse nucleoplasmic fraction of Pol II LS is relatively hypophosphorylated on the CTD, and it is easily extracted by detergents. In transcriptionally active nuclei, speckle bound hyperphosphorylated Pol II LS molecules are distributed in irregularly shaped speckle domains, which appear to be interconnected via a reticular network. When transcription is inhibited, hyperphosphorylated Pol II LS and splicing protein SC35 accumulate in speckle domains, which are transformed into enlarged, dot-like structures lacking interconnections. When cells are released from transcriptional inhibition, Pol IIO and SC35 redistribute back to the interconnected speckle pattern of transcriptionally active cells. The redistribution of Pol II and SC35 is synchronous, reversible, and temperature dependent. It is concluded that: (a) hyperphosphorylation of Pol II LS's CTD is a better indicator of its tight association to discrete subnuclear domains than its transcriptional activity; (b) during states of transcriptional inhibition, hyperphosphorylated Pol II LS can be stored in enlarged speckle domains, which under the light microscope appear to coincide with the storage sites for splicing proteins; and (c) Pol II and splicing proteins redistribute simultaneously according to the overall transcriptional activity of the nucleus.


Subject(s)
Cell Nucleus/enzymology , Nuclear Proteins/metabolism , RNA Polymerase II/metabolism , Ribonucleoproteins , Spliceosomes/metabolism , Transcription, Genetic , Amanitins/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Cell Line , Cell Nucleus/metabolism , Dichlororibofuranosylbenzimidazole/pharmacology , Epitopes/analysis , Liver/enzymology , Molecular Sequence Data , Phosphorylation , RNA Polymerase II/analysis , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/immunology , RNA Polymerase II/isolation & purification , Rats , Temperature , Transcription, Genetic/drug effects
14.
J Cell Sci ; 107 ( Pt 3): 387-96, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8006060

ABSTRACT

Cytostellin, a approximately 240 kDa phosphoprotein found in all cells examined from human to yeast, is predominantly intranuclear in interphase mammalian cells and undergoes continuous redistribution during the cell cycle. Here, mammalian cytostellin is shown to localize to intranuclear regions enriched with multiple splicing proteins, including spliceosome assembly factor, SC-35. Cytostellin and the splicing proteins also co-localize to discrete foci (called 'dots'), which are distributed throughout the cell during mitosis and part of G1. The cytostellin that is localized to these dots resists extraction by Triton X-100, indicating that it is tightly associated with insoluble cell structures. All immunostainable cytostellin reappears in the nucleus before S-phase. Although cytostellin and the splicing proteins co-localize in interphase and dividing cells, cytostellin is not detected in purified spliceosomes, and it associates with six unidentified proteins, forming a macromolecular complex that is biochemically distinct from the proteins that comprise spliceosomes. This macromolecular complex is detected at constant levels throughout the cell cycle, and the level of cytostellin protein remains constant during the cell cycle. Nevertheless, intranuclear cytostellin immunostaining fluctuates markedly during the cell cycle. The monoclonal antibody (mAb) H5 epitope of cytostellin is 'masked' in serum-starved cells, but 60 minutes after serum stimulation intense cytostellin immunoreactivity appears in the nuclear speckles. This rapid induction of cytostellin immunoreactivity in subnuclear regions enriched with many splicing factors, as well as accumulations of RNA polymerase II (Pol II) transcripts, suggests that cytostellin may have a function related to mRNA biogenesis.


Subject(s)
Cell Nucleus/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , RNA Splicing , Ribonucleoproteins , Animals , Cell Cycle , Cell Line , Dogs , Fluorescent Antibody Technique , Mitosis
15.
J Biol Chem ; 266(11): 7207-13, 1991 Apr 15.
Article in English | MEDLINE | ID: mdl-2016323

ABSTRACT

In mammalian brain, physiological signals carried by cAMP seem to be targeted to intraneuronal sites by the association of cAMP-dependent protein kinase II beta with anchoring proteins that bind the regulatory subunit (RII beta) of the enzyme. Previously, an RII beta-binding domain was characterized in a large (Mr approximately 150,000) candidate anchor protein, rat brain P150 (Bregman, D. B., Bhattacharyya, N., and Rubin, C. S. (1989) J. Biol. Chem. 264, 4648-4656). RII beta-binding proteins with Mr values of 65,000-80,000 were detected in the brains of other species. Since little was known about the structural features of these lower Mr proteins, we undertook the characterization of bovine brain P75 as a prototype. A cDNA encoding 258 amino acid residues at the C terminus of P75 was cloned by probing a lambda gt11 expression library with 32P-RII beta. The cDNA insert was ligated into the pET-3b expression plasmid, and large amounts of the partial P75 polypeptide (designated P47) were produced in Escherichia coli. A purification scheme that yielded 9 mg of soluble P47 from a 1-liter bacterial culture was devised. Antibodies directed against the P47 polypeptide revealed that P75 is expressed almost exclusively in brain. The sequence of 117 amino acid residues at the C terminus of P75 contains the RII beta-binding site and is 80% identical to the corresponding region of P150. In contrast, a lower level of identity (36%) between P75 and P150 at a more N-terminal region indicates that the two RII beta-binding proteins are related, but distinct proteins. P75 is not homologous to microtubule-associated protein 2, an RII alpha-selective binding protein, or any other previously studied proteins. C-terminal truncation analysis disclosed that the final 26 residues in P75 are essential for binding RII beta.


Subject(s)
Adaptor Proteins, Signal Transducing , Brain/metabolism , Carrier Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Kinases/metabolism , A Kinase Anchor Proteins , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/isolation & purification , Carrier Proteins/metabolism , Cattle , Cloning, Molecular , Escherichia coli/genetics , Gene Library , Macromolecular Substances , Molecular Sequence Data , Molecular Weight , Nerve Tissue Proteins/isolation & purification , Nerve Tissue Proteins/metabolism , Organ Specificity , RNA, Messenger/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid
16.
J Biol Chem ; 264(8): 4648-56, 1989 Mar 15.
Article in English | MEDLINE | ID: mdl-2538452

ABSTRACT

Cyclic AMP-dependent protein kinase II-B appears to be adapted for function in the mammalian central nervous system via the properties of its regulatory subunit (RII-B). RII-B is selectively expressed in the central nervous system, tightly associated with cerebral cortex membranes, and avidly complexed by the bovine brain calmodulin-binding protein designated P75 (Sarkar, D., Erlichman, J., and Rubin, C. S. (1984) J. Biol. Chem. 259, 9840-9846). Complexes of RII-B and P75 polypeptides can be purified to near homogeneity from either membrane or cytosolic fractions of brain homogenates, suggesting that the binding protein plays a role in determining the central nervous system-specific properties of protein kinase II-B. To investigate the properties of a prototypic, nonabundant, RII-B-binding protein, we have cloned and characterized cDNAs for rat brain P150, a homolog of bovine brain P75. cDNAs were retrieved from a lambda gt11 expression library using 32P-labeled RII-B as a functional probe. cDNA inserts (800 and 1100 base pairs) subcloned into expression plasmids directed the production of partial P150 polypeptides in Escherichia coli that bind RII-B. Sequence analyses disclosed that P150 is a previously uncharacterized protein that contains multiple octapeptide repeats as well as unique sequences. Antibodies directed against 15-residue peptides corresponding to either repeated or unique sequences bound the polypeptides expressed in E. coli and a 150-kDa protein in rat brain membranes and cytosol. Moreover, the immunoprecipitated 150-kDa protein exhibited high affinity RII-B-binding activity. Finally, 3' deletion analysis demonstrated that a 15-amino acid segment of P150 is essential for binding with RII-B.


Subject(s)
Adaptor Proteins, Signal Transducing , Brain Chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Protein Kinases/metabolism , A Kinase Anchor Proteins , Amino Acid Sequence , Animals , Base Sequence , Cattle , Cell Membrane/analysis , Cerebral Cortex/analysis , Cloning, Molecular , Cyclic AMP/pharmacology , Cytosol/analysis , DNA/genetics , Genetic Vectors , Isoenzymes/metabolism , Molecular Sequence Data , Plasmids , RNA, Messenger/genetics , Rabbits , Rats , Rats, Inbred Strains , Structure-Activity Relationship , Swine , Transformation, Bacterial
SELECTION OF CITATIONS
SEARCH DETAIL
...