Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters











Publication year range
1.
Biochem Biophys Res Commun ; 484(4): 864-870, 2017 03 18.
Article in English | MEDLINE | ID: mdl-28188792

ABSTRACT

Centrosomal protein of 192 kDa (CEP192) is a scaffolding protein that recruits the mitotic protein kinases Aurora A and PLK1 to the centrosome. Here we demonstrate that CEP192 also recruits the type one protein phosphatase (PP1) via a highly conserved KHVTF docking motif. The threonine of the KHVTF motif is phosphorylated during mitosis and protein kinase inhibition studies suggest this to be a PLK1-dependent process.


Subject(s)
Centrosome/metabolism , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , Mitosis/physiology , Receptors, Neuropeptide Y/chemistry , Receptors, Neuropeptide Y/metabolism , Binding Sites , Centrosome/ultrastructure , Enzyme Activation , HeLa Cells , Humans , Phosphorylation , Protein Binding
2.
Gynecol Oncol ; 145(1): 176-184, 2017 04.
Article in English | MEDLINE | ID: mdl-28131528

ABSTRACT

OBJECTIVE: The management of locally advanced cervical cancer has improved significantly with the advent of cisplatin-based chemoradiotherapy (CRT) as the primary treatment regimen. Nevertheless, a significant proportion of patients fail to respond or relapse on this treatment and have a very poor prognosis. Our goal was to determine the prognostic value of a panel of proteins involved in detection and repair of DNA damage. METHODS: We performed fluorescence immunohistochemistry, and used software analysis to assess expression of DNA damage response proteins ATM, DNA-PKcs, PARP-1, Ku70 and Ku86 in 117 pre-treatment specimens from patients with locally advanced cervical cancer. We compared expression to clinicopathologic correlates to determine prognostic significance. RESULTS: Five-year progression-free survival was significantly lower in the low expressors than in high expressors of ATM (35% vs. 58%, p=0.044) and PARP-1 (24% vs. 61%, p=0.003), and showed a trend to significance for DNA-PKcs (30% vs. 60%, p=0.050). Low expression of the same proteins also correlated significantly with lower overall survival. In multivariable analysis, adjusted for FIGO stage and tumor size, low ATM and PARP-1 expression was significantly associated with both poorer progression-free and overall survival. Pairwise analyses indicated that expression levels of these proteins were correlated. CONCLUSIONS: Expression of DNA damage response proteins in cervical cancer is associated with outcome in patients treated with CRT. Immunohistochemical analysis of these proteins may be useful in guiding treatment decisions in such patients.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy/methods , Cisplatin/therapeutic use , Uterine Cervical Neoplasms/therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , DNA Damage , DNA-Activated Protein Kinase/metabolism , Disease-Free Survival , Female , Fluorescence , Humans , Immunohistochemistry , Ku Autoantigen/metabolism , Middle Aged , Multivariate Analysis , Neoplasm Staging , Nuclear Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Prognosis , Radiotherapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Young Adult
3.
Oncogene ; 29(27): 3990-6, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20473328

ABSTRACT

Replication-competent oncolytic viruses hold great potential for the clinical treatment of many cancers. Importantly, many oncolytic virus candidates, such as reovirus and myxoma virus, preferentially infect cancer cells bearing abnormal cellular signaling pathways. Reovirus and myxoma virus are highly responsive to activated Ras and Akt signaling pathways, respectively, for their specificity for viral oncolysis. However, considering the complexity of cancer cell populations, it is possible that other tumor-specific signaling pathways may also contribute to viral discrimination between normal versus cancer cells. Because carcinogenesis is a multistep process involving the accumulation of both oncogene activations and the inactivation of tumor suppressor genes, we speculated that not only oncogenes but also tumor suppressor genes may have an important role in determining the tropism of these viruses for cancer cells. It has been previously shown that many cellular tumor suppressor genes, such as p53, ATM and Rb, are important for maintaining genomic stability; dysfunction of these tumor suppressors may disrupt intact cellular antiviral activity due to the accumulation of genomic instability or due to interference with apoptotic signaling. Therefore, we speculated that cells with dysfunctional tumor suppressors may display enhanced susceptibility to challenge with these oncolytic viruses, as previously seen with adenovirus. We report here that both reovirus and myxoma virus preferentially infect cancer cells bearing dysfunctional or deleted p53, ATM and Rb tumor suppressor genes compared to cells retaining normal counterparts of these genes. Thus, oncolysis by these viruses may be influenced by both oncogenic activation and tumor suppressor status.


Subject(s)
Genes, Tumor Suppressor , Myxoma virus/physiology , Neoplasms/genetics , Neoplasms/virology , Oncolytic Viruses/physiology , Reoviridae/physiology , Viral Tropism , Animals , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Deletion , Gene Knockdown Techniques , Humans , Mice , Neoplasms/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
4.
Biosens Bioelectron ; 20(5): 918-24, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15530788

ABSTRACT

In the development of biosensors, it is essential to understand how the signal-transducing element may perturb surface-bound proteins and nucleic acids. The tip of the atomic force microscope is such an element in atomic force microscopy. In this paper, we describe the influence of tip-sample interactions on the measured height of the DNA repair protein, Ku, that has been adsorbed onto a mica surface which was submerged in aqueous solution. We find that the measured height of the Ku molecule depends critically on whether or not it is associated with DNA. Additionally, we observed that the conditions (time and concentration) under which Ku is incubated with DNA, affect the appearance (number and type) of the DNA-Ku complexes observed.


Subject(s)
Biosensing Techniques/methods , DNA Helicases/chemistry , DNA Helicases/ultrastructure , Microscopy, Atomic Force/methods , Plasmids/chemistry , Plasmids/ultrastructure , Water/chemistry , Biosensing Techniques/instrumentation , Coated Materials, Biocompatible/analysis , Coated Materials, Biocompatible/chemistry , DNA Helicases/analysis , DNA-Binding Proteins/analysis , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/ultrastructure , Image Interpretation, Computer-Assisted/methods , Ku Autoantigen , Nucleic Acid Conformation , Plasmids/analysis , Protein Binding , Protein Conformation , Surface Properties , Water/analysis
5.
Biochimie ; 85(11): 1161-73, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14726021

ABSTRACT

DNA double strand breaks (DSB) are the most serious form of DNA damage. If not repaired they can lead to cell death. If misrepaired DSBs contribute to chromosomal aberrations and genomic instability. Non-homologous end joining (NHEJ) is one of two major pathways for the repair of DSBs in human cells. Proteins known to be required for NHEJ include the DNA-dependent protein kinase (DNA-PK), XRCC4, DNA ligase IV, and Artemis. This review discusses how these and other accessory proteins may function in the repair of DSBs produced by ionizing radiation (IR) and by V(D)J recombination.


Subject(s)
DNA Damage , DNA Repair , Recombination, Genetic/physiology , Animals , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Evolution, Molecular , Humans , Nuclear Proteins , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Protein Serine-Threonine Kinases/therapeutic use , Recombination, Genetic/genetics
6.
J Biol Chem ; 276(22): 18992-8, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11376007

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a complex of DNA-PK catalytic subunit (DNA-PKcs) and the DNA end-binding Ku70/Ku80 heterodimer. DNA-PK is required for DNA double strand break repair by the process of nonhomologous end joining. Nonhomologous end joining is a major mechanism for the repair of DNA double strand breaks in mammalian cells. As such, DNA-PK plays essential roles in the cellular response to ionizing radiation and in V(D)J recombination. In vitro, DNA-PK undergoes phosphorylation of all three protein subunits (DNA-PK catalytic subunit, Ku70 and Ku80) and phosphorylation correlates with inactivation of the serine/threonine protein kinase activity of DNA-PK. Here we show that phosphorylation-induced loss of the protein kinase activity of DNA-PK is restored by the addition of the purified catalytic subunit of either protein phosphatase 1 or protein phosphatase 2A (PP2A) and that this reactivation is blocked by the potent protein phosphatase inhibitor, microcystin. We also show that treating human lymphoblastoid cells with either okadaic acid or fostriecin, at PP2A-selective concentrations, causes a 50-60% decrease in DNA-PK protein kinase activity, although the protein phosphatase 1 activity in these cells was unaffected. In vivo phosphorylation of DNA-PKcs, Ku70, and Ku80 was observed when cells were labeled with [(32)P]inorganic phosphate in the presence of the protein phosphatase inhibitor, okadaic acid. Together, our data suggest that reversible protein phosphorylation is an important mechanism for the regulation of DNA-PK protein kinase activity and that the protein phosphatase responsible for reactivation in vivo is a PP2A-like enzyme.


Subject(s)
Antigens, Nuclear , DNA Helicases , Protein Serine-Threonine Kinases/metabolism , Alkenes/pharmacology , Catalysis , Catalytic Domain , Cell Line , Cells, Cultured , DNA Damage , DNA Repair , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Ku Autoantigen , Microcystins , Nuclear Proteins/metabolism , Okadaic Acid/pharmacology , Peptides, Cyclic/pharmacology , Phosphates/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Polyenes , Precipitin Tests , Protein Binding , Protein Kinases/metabolism , Protein Phosphatase 1 , Protein Phosphatase 2 , Pyrones , Recombinant Proteins/metabolism , Time Factors
7.
J Biol Chem ; 276(26): 24323-30, 2001 Jun 29.
Article in English | MEDLINE | ID: mdl-11309379

ABSTRACT

To examine determinants of fidelity in DNA end joining, a substrate containing a model of a staggered free radical-mediated double-strand break, with cohesive phosphoglycolate-terminated 3'-overhangs and a one-base gap in each strand, was constructed. In extracts of Xenopus eggs, human lymphoblastoid cells, hamster CHO-K1 cells, and a Chinese hamster ovary (CHO) derivative lacking the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), the predominant end joining product was that corresponding to accurate restoration of the original sequence. In extracts of the Ku-deficient CHO derivative xrs6, a shorter product, consistent with 3' --> 5' resection before ligation, was formed. Similar results were seen for a substrate with 5'-overhangs and recessed 3'-phosphoglycolate ends. Supplementation of the xrs6 extracts with purified Ku restored accurate end joining. In Xenopus and human extracts, but not in hamster extracts, gap filling and ligation were blocked by wortmannin, consistent with a requirement for DNA-PKcs activity. The results suggest a Ku-dependent pathway, regulated by DNA-PKcs, that can accurately restore the original DNA sequence at sites of free radical-mediated double-strand breaks, by protecting DNA termini from degradation and maintaining the alignment of short partial complementarities during gap filling and ligation.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA Repair , DNA-Binding Proteins/physiology , Glycolates/metabolism , Nuclear Proteins/physiology , Androstadienes/pharmacology , Animals , CHO Cells , Cell Extracts/analysis , Cricetinae , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , Enzyme Inhibitors/pharmacology , Humans , Ku Autoantigen , Mutation , Nuclear Proteins/genetics , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , Oocytes/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Species Specificity , Wortmannin , Xenopus
8.
J Biol Chem ; 276(21): 17914-9, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11279124

ABSTRACT

Cells can respond to DNA damage by activating checkpoints that delay cell cycle progression and allow time for DNA repair. Chemical inhibitors of the G(2) phase DNA damage checkpoint may be used as tools to understand better how the checkpoint is regulated and may be used to sensitize cancer cells to DNA-damaging therapies. However, few inhibitors are known. We used a cell-based assay to screen natural extracts for G(2) checkpoint inhibitors and identified debromohymenialdisine (DBH) from a marine sponge. DBH is distinct structurally from previously known G(2) checkpoint inhibitors. It inhibited the G(2) checkpoint with an IC(50) of 8 micrometer and showed moderate cytotoxicity (IC(50) = 25 micrometer) toward MCF-7 cells. DBH inhibited the checkpoint kinases Chk1 (IC(50) = 3 micrometer) and Chk2 (IC(50) = 3.5 micrometer) but not ataxia-telangiectasia mutated (ATM), ATM-Rad3-related protein, or DNA-dependent protein kinase in vitro, indicating that it blocks two major branches of the checkpoint pathway downstream of ATM. It did not cause the activation or inhibition of different signal transduction proteins, as determined by mobility shift analysis in Western blots, suggesting that it inhibits a narrow range of protein kinases in vivo.


Subject(s)
Azepines/pharmacology , DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , G2 Phase/drug effects , Protein Kinase Inhibitors , Protein Kinases , Protein Serine-Threonine Kinases , Pyrroles/pharmacology , Alkaloids/pharmacology , Animals , Cell Line , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Porifera , Signal Transduction/drug effects
9.
J Biol Chem ; 276(7): 4828-33, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11096068

ABSTRACT

Genistein is an isoflavenoid that is abundant in soy beans. Genistein has been reported to have a wide range of biological activities and to play a role in the diminished incidence of breast cancer in populations that consume a soy-rich diet. Genistein was originally identified as an inhibitor of tyrosine kinases; however, it also inhibits topoisomerase II by stabilizing the covalent DNA cleavage complex, an event predicted to cause DNA damage. The topoisomerase II inhibitor etoposide acts in a similar manner. Here we show that genistein induces the up-regulation of p53 protein, phosphorylation of p53 at serine 15, activation of the sequence-specific DNA binding properties of p53, and phosphorylation of the hCds1/Chk2 protein kinase at threonine 68. Phosphorylation and activation of p53 and phosphorylation of Chk2 were not observed in ATM-deficient cells. In contrast, the topoisomerase II inhibitor etoposide induced phosphorylation of p53 and Chk2 in ATM-positive and ATM-deficient cells. In addition, genistein-treated ATM-deficient cells were significantly more susceptible to genistein-induced killing than were ATM-positive cells. Together our data suggest that ATM is required for activation of a DNA damage-induced pathway that activates p53 and Chk2 in response to genistein.


Subject(s)
Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , Tumor Suppressor Protein p53/metabolism , Androstadienes/pharmacology , Ataxia Telangiectasia Mutated Proteins , Benzoquinones , Caffeine/pharmacology , Cell Cycle Proteins , Cell Line , Checkpoint Kinase 2 , DNA Fragmentation , DNA-Binding Proteins , Etoposide/pharmacology , Humans , Lactams, Macrocyclic , Mutation , Phosphorylation , Plants, Medicinal , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinones/pharmacology , Rifabutin/analogs & derivatives , Topoisomerase II Inhibitors , Tumor Suppressor Proteins , Wortmannin
11.
J Biol Chem ; 275(30): 22719-27, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10801797

ABSTRACT

The ataxia telangiectasia mutated (ATM) gene encodes a serine/threonine protein kinase that plays a critical role in genomic surveillance and development. Here, we use a peptide library approach to define the in vitro substrate specificity of ATM kinase activity. The peptide library analysis identified an optimal sequence with a central core motif of LSQE that is preferentially phosphorylated by ATM. The contributions of the amino acids surrounding serine in the LSQE motif were assessed by utilizing specific peptide libraries or individual peptide substrates. All amino acids comprising the LSQE sequence were critical for maximum peptide substrate suitability for ATM. The DNA-dependent protein kinase (DNA-PK), a Ser/Thr kinase related to ATM and important in DNA repair, was compared with ATM in terms of peptide substrate selectivity. DNA-PK was found to be unique in its preference of neighboring amino acids to the phosphorylated serine. Peptide library analyses defined a preferred amino acid motif for ATM that permits clear distinctions between ATM and DNA-PK kinase activity. Data base searches using the library-derived ATM sequence identified previously characterized substrates of ATM, as well as novel candidate substrate targets that may function downstream in ATM-directed signaling pathways.


Subject(s)
Peptide Library , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Substrate Specificity , Tumor Cells, Cultured , Tumor Suppressor Proteins
12.
Mol Cell Biol ; 20(11): 3807-16, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10805724

ABSTRACT

Three Saccharomyces cerevisiae proteins (Yng1/YOR064c, Yng2/YHR090c, and Pho23) and two Schizosaccharomyces pombe proteins (Png1/CAA15917 and Png2/CAA21250) share significant sequence identity with the human candidate tumor suppressor p33(ING1) in their C-terminal regions. The homologous regions contain PHD finger domains which have been implicated in chromatin-mediated transcriptional regulation. We show that GFP-Yng2, like human Ing1, is localized in the nucleus. Deletion of YNG2 results in several phenotypes, including an abnormal multibudded morphology, an inability to utilize nonfermentable carbon sources, heat shock sensitivity, slow growth, temperature sensitivity, and sensitivity to caffeine. These phenotypes are suppressed by expression of either human Ing1 or S. pombe Png1, suggesting that the yeast and human proteins are functionally conserved. Yng1- and Pho23-deficient cells also share some of these phenotypes. We demonstrated by yeast two-hybrid and coimmunoprecipitation tests that Yng2 interacts with Tra1, a component of histone acetyltransferase (HAT) complexes. We further demonstrated by coimmunoprecipitation that HA-Yng1, HA-Yng2, HA-Pho23, and HA-Ing1 are associated with HAT activities in yeast. Genetic and biochemical evidence indicate that the Yng2-associated HAT is Esa1, suggesting that Yng2 is a component of the NuA4 HAT complex. These studies suggest that the yeast Ing1-related proteins are involved in chromatin remodeling. They further suggest that these functions may be conserved in mammals and provide a possible mechanism for the human Ing1 candidate tumor suppressor.


Subject(s)
Acetyltransferases/metabolism , Fungal Proteins/metabolism , Genes, Tumor Suppressor , Membrane Proteins , Phospholipid Transfer Proteins , Proteins/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/enzymology , Schizosaccharomyces/enzymology , Amino Acid Sequence , Cell Cycle Proteins , Cell Nucleus/metabolism , DNA-Binding Proteins , Fungal Proteins/genetics , Fungal Proteins/physiology , Gene Expression , Genetic Complementation Test , Histone Acetyltransferases , Humans , Inhibitor of Growth Protein 1 , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutagenesis , Nuclear Proteins , Phenotype , Proteins/metabolism , Saccharomyces cerevisiae/genetics , Schizosaccharomyces/metabolism , Sequence Homology, Amino Acid , Tumor Suppressor Proteins
13.
J Biol Chem ; 275(11): 7803-10, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10713094

ABSTRACT

ATM is mutated in the human genetic disorder ataxia telangiectasia, which is characterized by ataxia, immune defects, and cancer predisposition. Cells that lack ATM exhibit delayed up-regulation of p53 in response to ionizing radiation. Serine 15 of p53 is phosphorylated in vivo in response to ionizing radiation, and antibodies to ATM immunoprecipitate a protein kinase activity that, in the presence of manganese, phosphorylates p53 at serine 15. Immunoprecipitates of ATM also phosphorylate PHAS-I in a manganese-dependent manner. Here we have purified ATM from human cells using nine chromatographic steps. Highly purified ATM phosphorylated PHAS-I, the 32-kDa subunit of RPA, serine 15 of p53, and Chk2 in vitro. The majority of the ATM phosphorylation sites in Chk2 were located in the amino-terminal 57 amino acids. In each case, phosphorylation was strictly dependent on manganese. ATM protein kinase activity was inhibited by wortmannin with an IC(50) of approximately 100 nM. Phosphorylation of RPA, but not p53, Chk2, or PHAS-I, was stimulated by DNA. The related protein, DNA-dependent protein kinase catalytic subunit, also phosphorylated PHAS-I, RPA, and Chk2 in the presence of manganese, suggesting that the requirement for manganese is a characteristic of this class of enzyme.


Subject(s)
Ataxia Telangiectasia/enzymology , Placenta/enzymology , Protein Serine-Threonine Kinases/isolation & purification , Androstadienes/pharmacology , Antibody Specificity , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Checkpoint Kinase 2 , Cross Reactions , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Female , Humans , Manganese , Nuclear Proteins , Phosphorylation , Pregnancy , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Replication Protein A , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , Wortmannin
14.
Radiat Res ; 151(4): 414-22, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10190493

ABSTRACT

The DNA-dependent protein kinase (DNA-PK) is composed of a large catalytic subunit (DNA-PKcs) and a DNA-binding protein, Ku. Cells lacking DNA-PK activity are radiosensitive and are defective in DNA double-strand break repair and V(D)J recombination. Although much information regarding the interactions of Ku with DNA ends is available, relatively little is known about the interaction of DNA-PKcs with DNA-bound Ku. Here we show, using electrophoretic mobility shift assays, that chemical crosslinkers enhance the formation of protein-DNA complexes containing DNA-PKcs, Ku and other proteins in extracts from cells of normal human cell lines. Extracts from cells of the radiosensitive human cell line M059J, which lacks DNA-PKcs, are not competent to form these protein-DNA complexes, while addition of purified DNA-PKcs protein restores complex formation. This assay may be useful for screening for DNA-PK function in cells of human cell lines and for identifying proteins that interact with the DNA-PK-DNA complex. We also show that Ku protein in rodent cells can interact with human DNA-PKcs; however, this assay may be less useful for studying Ku/DNA-PKcs interactions in cells of rodent cell lines due to the low abundance of DNA-PKcs in these cells.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , 3T3 Cells , Animals , DNA-Activated Protein Kinase , HeLa Cells , Humans , Ku Autoantigen , Mice
15.
Biochim Biophys Acta ; 1430(1): 119-26, 1999 Feb 10.
Article in English | MEDLINE | ID: mdl-10082940

ABSTRACT

Poly(ADP-ribose) polymerase (PARP) and DNA-dependent protein kinase (DNA-PK) are important nuclear enzymes that cooperate to minimize genomic damage caused by DNA strand interruptions. DNA strand interruptions trigger the ADP-ribosylation activity and phosphorylation activity of PARP and DNA-PK respectively. In order to understand the relationship of PARP and DNA-PK with respect to DNA binding required for their activation, we analyzed the kinetics of the reactions and determined the apparent dissociation constants (Kd app) of the enzymes for DNA strand interruptions. PARP has a high binding affinity for blunt ends of DNA (Kd app=116 pM) and 3' single-base overhangs (Kd app=332 pM) in comparison to long overhangs (Kd app=2.6-5.0 nM). Nicks are good activators of PARP although the affinity of PARP for nicks (Kd app=467 pM) is 4-fold less than that for blunt ends. The Kd app of DNA-PK for 3' single-base overhangs, blunt ends and long overhangs is 704 pM, 1.3 nM and 1.4-2.2 nM respectively. These results demonstrate that (1) PARP, when compared to DNA-PK, has a greater preference for blunt ends and 3' single-base overhangs but a weaker preference for long overhangs, and (2) nicks are effective in attracting and activating PARP. The possible implications of the preferences of PARP and DNA-PK for DNA strand interruptions in vivo are discussed.


Subject(s)
DNA Damage , DNA-Binding Proteins , Plasmids/chemistry , Poly(ADP-ribose) Polymerases/chemistry , Protein Serine-Threonine Kinases/chemistry , DNA-Activated Protein Kinase , Enzyme Activation , Escherichia coli/metabolism , Poly(ADP-ribose) Polymerases/biosynthesis , Poly(ADP-ribose) Polymerases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics
16.
Biochemistry ; 38(6): 1819-28, 1999 Feb 09.
Article in English | MEDLINE | ID: mdl-10026262

ABSTRACT

Ku antigen is composed of 70 and 82 kDa subunits (Ku70 and Ku80, respectively) that together bind with high affinity to ends of double-stranded DNA and other DNA structures in vitro. When bound to DNA, the Ku 70/80 heterodimer enhances the kinase activity of the catalytic subunit of the DNA-dependent protein kinase, DNA-PKcs. Ku and DNA-PKcs are required for V(D)J recombination and DNA double-strand break repair in vivo and may also play a role in regulation of transcription. Ku is phosphorylated by DNA-PKcs in vitro, and cells that lack DNA-PKcs are deficient in Ku phosphorylation in vitro, suggesting that Ku may be a physiological target for DNA-PK. Here we have identified the sites of DNA-PK phosphorylation in human Ku protein. We find that Ku70 is phosphorylated at a single serine residue, serine 6, located in the putative transcriptional activation domain, and Ku80 is phosphorylated at serines 577 and 580 and at threonine 715. Interestingly, none of the phosphorylation sites identified in Ku correspond to the serine-glutamine consensus for DNA-PK phosphorylation, consistent with previous reports that DNA-PK can recognize additional phosphorylation motifs.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cricetinae , DNA-Activated Protein Kinase , DNA-Binding Proteins/isolation & purification , Dimerization , Humans , Ku Autoantigen , Mice , Molecular Sequence Data , Molecular Weight , Nuclear Proteins/isolation & purification , Phosphopeptides/isolation & purification , Phosphopeptides/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/isolation & purification , Rats , Sequence Alignment
17.
J Virol ; 73(1): 650-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9847370

ABSTRACT

Herpes simplex virus type 1 (HSV-1) infection causes the active degradation of the catalytic subunit of DNA-dependent protein kinase (DNA-PKcs), and this process is reliant on the expression of the HSV-1 immediate-early protein Vmw110. In this study we investigated in more detail the mechanism by which the degradation occurs, the domains of Vmw110 which are required, and whether Vmw110 is by itself sufficient for the effect. We found that proteasome inhibitors prevented the degradation of DNA-PKcs, indicating the involvement of a proteasome pathway. Furthermore, the continued activity of DNA-PK during infection in the presence of these inhibitors indicated that Vmw110 does not directly alter the enzyme activity of DNA-PKcs prior to its degradation in a normal infection. Indeed, Vmw110 was found to bind to neither the catalytic nor Ku subunits of DNA-PK. Using mutant Vmw110 viruses we show that the RING finger domain of Vmw110 is essential for the induced degradation of DNA-PKcs but that the ability of Vmw110 to bind to a cellular ubiquitin-specific protease (HAUSP) is not required. When expressed in the absence of other viral proteins, Vmw110 was sufficient to cause the degradation of DNA-PKcs, indicating that the effect on the stability of DNA-PKcs was a direct consequence of Vmw110 activity and not an indirect Vmw110-dependent effect of virus infection. Finally, the Vmw110-induced degradation of DNA-PKcs and loss in DNA-PK activity appears to be beneficial to HSV-1 infection, as virus replication was more efficient in cells lacking DNA-PKcs, especially at low multiplicities of infection.


Subject(s)
Cysteine Endopeptidases/physiology , DNA-Binding Proteins , Herpesvirus 1, Human/physiology , Immediate-Early Proteins/physiology , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/metabolism , Binding Sites , DNA-Activated Protein Kinase , HeLa Cells , Humans , Nuclear Proteins , Proteasome Endopeptidase Complex , Ubiquitin-Protein Ligases , Virus Replication
18.
Nat Genet ; 20(4): 398-400, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9843217

ABSTRACT

The human genetic disorder ataxia-telangiectasia (AT) is characterized by immunodeficiency, progressive cerebellar ataxia, radiosensitivity, cell cycle checkpoint defects and cancer predisposition. The gene mutated in this syndrome, ATM (for AT mutated), encodes a protein containing a phosphatidyl-inositol 3-kinase (PI-3 kinase)-like domain. ATM also contains a proline-rich region and a leucine zipper, both of which implicate this protein in signal transduction. The proline-rich region has been shown to bind to the SH3 domain of c-Abl, which facilitates its phosphorylation and activation by ATM. Previous results have demonstrated that AT cells are defective in the G1/S checkpoint activated after radiation damage and that this defect is attributable to a defective p53 signal transduction pathway. We report here direct interaction between ATM and p53 involving two regions in ATM, one at the amino terminus and the other at the carboxy terminus, corresponding to the PI-3 kinase domain. Recombinant ATM protein phosphorylates p53 on serine 15 near the N terminus. Furthermore, ectopic expression of ATM in AT cells restores normal ionizing radiation (IR)-induced phosphorylation of p53, whereas expression of ATM antisense RNA in control cells abrogates the rapid IR-induced phosphorylation of p53 on serine 15. These results demonstrate that ATM can bind p53 directly and is responsible for its serine 15 phosphorylation, thereby contributing to the activation and stabilization of p53 during the IR-induced DNA damage response.


Subject(s)
Protein Serine-Threonine Kinases , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ataxia Telangiectasia Mutated Proteins , Binding Sites , Cell Cycle Proteins , DNA-Binding Proteins , Humans , Phosphorylation , Protein Binding , Proteins/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Proteins
19.
Mol Cell Biol ; 18(11): 6719-28, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9774685

ABSTRACT

Protein kinase Cdelta (PKCdelta) is proteolytically cleaved and activated at the onset of apoptosis induced by DNA-damaging agents, tumor necrosis factor, and anti-Fas antibody. A role for PKCdelta in apoptosis is supported by the finding that overexpression of the catalytic fragment of PKCdelta (PKCdelta CF) in cells is associated with the appearance of certain characteristics of apoptosis. However, the functional relationship between PKCdelta cleavage and induction of apoptosis is unknown. The present studies demonstrate that PKCdelta associates constitutively with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs). The results show that PKCdelta CF phosphorylates DNA-PKcs in vitro. Interaction of DNA-PKcs with PKCdelta CF inhibits the function of DNA-PKcs to form complexes with DNA and to phosphorylate its downstream target, p53. The results also demonstrate that cells deficient in DNA-PK are resistant to apoptosis induced by overexpressing PKCdelta CF. These findings support the hypothesis that functional interactions between PKCdelta and DNA-PK contribute to DNA damage-induced apoptosis.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C/metabolism , Protein Serine-Threonine Kinases/metabolism , Apoptosis/physiology , Binding Sites/physiology , Caspase 3 , Caspases/metabolism , Cell Line , DNA Damage/genetics , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Humans , Microscopy, Fluorescence , Nuclear Proteins , Peptide Fragments/metabolism , Phosphorylation , Protein Binding/physiology , Protein Kinase C-delta , Transfection/genetics , Tumor Suppressor Protein p53/metabolism
20.
J Biol Chem ; 273(41): 26559-65, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9756893

ABSTRACT

The yeast Ada and TBP class of Spt proteins interact in multiple complexes that are required for transcriptional regulation. We have identified Tra1p as a component of these complexes through tandem mass spectrometry analysis of proteins that associate with Ngg1p/Ada3p. TRA1 is an essential gene and encodes a 3744-amino acid protein that is a member of a group of proteins including the catalytic subunit of DNA-dependent protein kinase, ATM and TRRAP, with carboxyl-terminal regions related to phosphatidylinositol 3-kinases. The interaction between Tra1p and Ada/Spt components was verified by the reciprocal coimmunoprecipitation of Ada2p and Tra1p from whole cell extracts in one or more complexes containing Spt7p. Tra1p cofractionated with Ngg1p and Spt7p through consecutive chromatography on Mono Q, DNA-cellulose, and Superose 6 columns. Binding of Tra1p to DNA-cellulose required Ada components. The association of Tra1p with two Ada.Spt complexes was suggested by its cofractionation with Ngg1p and Spt7p in two peaks on the Mono Q column. In the absence of Ada2p, the elution profile of Tra1p shifted to a distinct peak. Despite the similarity of Tra1p to a group of putative protein kinases, we have not detected protein kinase activity within immunoprecipitates of Tra1p or the Ada.Spt complexes.


Subject(s)
Carrier Proteins , Cell Cycle Proteins , DNA-Binding Proteins , Gene Expression Regulation, Fungal , Proteins/metabolism , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , E2F Transcription Factors , Histone Acetyltransferases , Molecular Sequence Data , Precipitin Tests , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Retinoblastoma-Binding Protein 1 , Transcription Factor DP1
SELECTION OF CITATIONS
SEARCH DETAIL