Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 13469, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34188089

ABSTRACT

G-quadruplexes (G4) are non-canonical secondary structures consisting in stacked tetrads of hydrogen-bonded guanines bases. An essential feature of G4 is their intrinsic polymorphic nature, which is characterized by the equilibrium between several conformations (also called topologies) and the presence of different types of loops with variable lengths. In cells, G4 functions rely on protein or enzymatic factors that recognize and promote or resolve these structures. In order to characterize new G4-dependent mechanisms, extensive researches aimed at identifying new G4 binding proteins. Using G-rich single-stranded oligonucleotides that adopt non-controlled G4 conformations, a large number of G4-binding proteins have been identified in vitro, but their specificity towards G4 topology remained unknown. Constrained G4 structures are biomolecular objects based on the use of a rigid cyclic peptide scaffold as a template for directing the intramolecular assembly of the anchored oligonucleotides into a single and stabilized G4 topology. Here, using various constrained RNA or DNA G4 as baits in human cell extracts, we establish the topology preference of several well-known G4-interacting factors. Moreover, we identify new G4-interacting proteins such as the NELF complex involved in the RNA-Pol II pausing mechanism, and we show that it impacts the clastogenic effect of the G4-ligand pyridostatin.


Subject(s)
DNA-Binding Proteins/chemistry , G-Quadruplexes , Oligonucleotides/chemistry , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Transcription Factors/chemistry
2.
Pathol Biol (Paris) ; 54(4): 185-93, 2006 May.
Article in English | MEDLINE | ID: mdl-16563661

ABSTRACT

A line of investigation in the search for sensitizing tumor cells to chemotherapy or radiotherapy relies on the selection of DNA repair inhibitors. In the area of DNA repair mechanisms, DNA-dependent protein kinase (DNA-PK) represents a key complex. Indeed DNA-PK is involved in the non-homologous end joining (NHEJ) process that corresponds to the major activity responsible for cell survival after ionizing radiation or chemotherapeutic treatment producing DNA double strand breaks. DNA-PK belongs to the PI3-K related kinase family and specific inhibitors have been recently selected and evaluated as radio- and chemo-sensitizers. These drugs, along with other ways to inhibit the DSBs repair process, are presented and discussed.


Subject(s)
Calcium-Binding Proteins/drug effects , DNA Damage , DNA Repair , DNA-Activated Protein Kinase/drug effects , DNA-Activated Protein Kinase/radiation effects , Neoplasms/drug therapy , Neoplasms/radiotherapy , Calcium-Binding Proteins/radiation effects , Humans
3.
Oncogene ; 20(32): 4373-82, 2001 Jul 19.
Article in English | MEDLINE | ID: mdl-11466618

ABSTRACT

The heterodimeric Ku protein (composed of the Ku 86 and Ku 70 sub-units) is a nuclear protein which binds to DNA termini without sequence specificity. Ku is the DNA-targeting component of the large catalytic sub-unit of the DNA-dependent protein kinase complex that is required for the repair of DNA double-strand breaks in mammalian cells. We studied the expression and function of Ku/DNA-PK during granulocytic differentiation of two human promyelocytic cell lines, HL60 and NB4, a process associated to decreased radiation resistance. After 3 days exposure to differentiating agents (either all-trans-retinoic acid or DMSO), Ku binding to double stranded (ds)-DNA ends declined dramatically whereas Ku protein levels remain unchanged. The nuclear, but not cytoplasmic, fraction of differentiated HL60 cells extracts exhibited a heat-sensitive inhibitory activity towards DNA binding of recombinant Ku heterodimer. We further demonstrate that immunoprecipitation of Ku is impaired in extracts from differentiated cells by using two antibodies that recognize epitopes within the C-terminus DNA binding domains of Ku 70 and Ku 86 proteins. These results favor the hypothesis of a protein interacting with Ku that would prevent DNA binding of heterodimerized Ku protein by steric hindrance.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/metabolism , Granulocytes/physiology , Myeloid Progenitor Cells/physiology , Nuclear Proteins/metabolism , Antibodies, Monoclonal/immunology , Cell Differentiation , Cell Extracts/analysis , Cell Line , Cell Nucleus/metabolism , DNA Repair , DNA-Binding Proteins/immunology , DNA-Binding Proteins/physiology , Dimerization , Down-Regulation , Epitopes/immunology , HL-60 Cells , Humans , Ku Autoantigen , Nuclear Proteins/immunology , Nuclear Proteins/physiology , Precipitin Tests , Protein Structure, Tertiary
4.
FASEB J ; 14(12): 1765-74, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973926

ABSTRACT

The nucleotide excision repair pathway contributes to genetic stability by removing a wide range of DNA damage through an error-free reaction. When the lesion is located, the altered strand is incised on both sides of the lesion and a damaged oligonucleotide excised. A repair patch is then synthesized and the repaired strand is ligated. It is assumed that only DNA polymerases delta and/or epsilon participate to the repair DNA synthesis step. Using UV and cisplatin-modified DNA templates, we measured in vitro that extracts from cells overexpressing the error-prone DNA polymerase beta exhibited a five- to sixfold increase of the ultimate DNA synthesis activity compared with control extracts and demonstrated the specific involvement of Pol beta in this step. By using a 28 nt gapped, double-stranded DNA substrate mimicking the product of the incision step, we showed that Pol beta is able to catalyze strand displacement downstream of the gap. We discuss these data within the scope of a hypothesis previously presented proposing that excess error-prone Pol beta in cancer cells could perturb the well-defined specific functions of DNA polymerases during error-free DNA transactions.


Subject(s)
DNA Polymerase beta/physiology , DNA Repair/physiology , DNA, Neoplasm/genetics , Animals , Base Sequence , CHO Cells , Cell Extracts , Cricetinae , DNA, Neoplasm/analysis , Humans , Molecular Sequence Data , Mutagenesis , Tumor Cells, Cultured
5.
Br J Cancer ; 83(4): 514-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10945500

ABSTRACT

Low-dose hyper-radiosensitivity (HRS) (below 0.5 Gy) has been extensively documented in the past few years. The molecular basis of this phenomenon remains largely unknown and the purpose of this study was to investigate the possible implication of the DNA repair DNA-PK complex. The activity of the DNA-PK complex, i.e. Ku DNA-end binding activity and kinase activity of the whole complex, was studied in 10 human cancer cell lines, 2 h after 0.2, 0.5 and 1 Gy irradiation. After low-dose irradiation (0.2 Gy), a marked decrease in DNA-PK activity was found in all six cell lines exhibiting HRS, whereas the DNA-PK activity was increased in the four cell lines which did not exhibit HRS. This modulation of DNA-PK activity was a rapid phenomenon occurring within the 2 h following low-dose radiation exposure. These data strongly suggest the implication of the DNA-PK repair complex in the HRS phenomenon.


Subject(s)
DNA-Binding Proteins , Protein Serine-Threonine Kinases/metabolism , Radiation Tolerance/physiology , Tumor Cells, Cultured/radiation effects , Amino Acid Sequence , Blotting, Western , DNA Repair/physiology , DNA Repair/radiation effects , DNA-Activated Protein Kinase , Dose-Response Relationship, Radiation , Humans , Molecular Sequence Data , Nuclear Proteins , Protein Serine-Threonine Kinases/radiation effects , Tumor Cells, Cultured/enzymology
6.
J Biol Chem ; 275(46): 35684-91, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10945984

ABSTRACT

Association of the DNA end-binding Ku70/Ku80 heterodimer with the 460-kDa serine/threonine kinase catalytic subunit forms the DNA-dependent protein kinase (DNA-PK) that is required for double-strand break repair by non-homologous recombination in mammalian cells. Recently, we have proposed a model in which the kinase activity is required for translocation of the DNA end-binding subunit Ku along the DNA helix when DNA-PK assembles on DNA ends. Here, we have questioned the consequences of Ku entry into DNA on local DNA processes by using human nuclear cell extracts incubated in the presence of linearized plasmid DNA. As two model processes, we have chosen nucleotide excision repair (NER) of UVC DNA lesions and transcription from viral promoters. We show that although NER efficiency is strongly reduced on linear DNA, it can be fully restored in the presence of DNA-PK inhibitors. Simultaneously, the amount of NER proteins bound to the UVC-damaged linear DNA is increased and the amount of Ku bound to the same DNA molecules is decreased. Similarly, the poor transcription efficiency exhibited by viral promoters on linear DNA is enhanced in the presence of DNA-PK inhibitor concentrations that prevent Ku entry into the DNA substrate molecule. The present results show that DNA-PK catalytic activity can regulate DNA transactions including transcription in the vicinity of double-strand breaks by controlling Ku entry into DNA.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Androstadienes/pharmacology , Base Sequence , DNA/chemistry , DNA/genetics , DNA Damage , DNA Repair/drug effects , DNA, Viral/chemistry , DNA, Viral/genetics , DNA, Viral/metabolism , DNA-Activated Protein Kinase , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Ku Autoantigen , Molecular Sequence Data , Nucleic Acid Conformation , Plasmids/chemistry , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Transcription, Genetic/drug effects , Ultraviolet Rays , Wortmannin
7.
Cancer Gene Ther ; 7(2): 339-46, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10770645

ABSTRACT

Ku86 has been shown to be involved in DNA double-strand break (DSB) repair and radiosensitivity in rodents, but its role in human cells is still under investigation. The purpose of this study was to evaluate the radiosensitivity and DSB repair after transfection of a Ku86-antisense in a human fibroblast cell line. Simian virus 40-transformed MRC5V1 human fibroblasts were transfected with a vector (pcDNA3) containing a Ku86-antisense cDNA. The main endpoints were Ku86 protein level, Ku DNA end-binding and DNA protein kinase activity, clonogenic survival, and DSB repair kinetics. After transfection of the Ku86-antisense, decreased Ku86 protein expression, Ku DNA end-binding activity, and DNA protein kinase activity were observed in the uncloned cellular population. The fibroblasts transfected with the Ku86-antisense showed also a radiosensitive phenotype, with a surviving fraction at 2 Gy of 0.29 compared with 0.75 for the control and 20% of unrepaired DSB observed at 24 hours after irradiation compared with 0% for the control. Several clones were also isolated with a decreased level of Ku86 protein, a surviving fraction at 2 Gy between 0.05 and 0.40, and 10-20% of unrepaired DSB at 24 hours. This study is the first to show the implication of Ku86 in DSB repair and in the radiosensitivity of human cells. This investigation strongly suggests that Ku86 could constitute an appealing target for combining gene therapy and radiation therapy.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Transfer Techniques , Nuclear Proteins/genetics , RNA, Antisense/genetics , Radiation Tolerance , Amino Acid Sequence , Blotting, Western , Cell Line , Cell Survival/radiation effects , Cesium Radioisotopes , Clone Cells/enzymology , Clone Cells/metabolism , Clone Cells/radiation effects , DNA Repair/radiation effects , DNA-Activated Protein Kinase , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , Fibroblasts/enzymology , Gamma Rays , Humans , Kinetics , Ku Autoantigen , Micronucleus Tests , Molecular Sequence Data , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/biosynthesis , Protein Serine-Threonine Kinases/metabolism , RNA, Antisense/radiation effects
8.
Arch Biochem Biophys ; 376(1): 26-33, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10729187

ABSTRACT

Reactive oxygen species (ROS) damage DNA which appears to represent the major target involved in mutagenesis, carcinogenesis, and aging cell responses. Various DNA modifications are generated by ROS, but 8-hydroxy-2'-deoxyguanosine (8-oxoG) has retained a lot of attention in the last few years. Therefore, numerous methods have been developed to detect and quantify the extent of 8-oxoG in DNA, most of them requiring a significant amount of DNA that might be limiting in the case of biological samples. 8-oxoG is repaired in Escherichia coli by a specific glycosylase, the Fpg (formamidopyrimidine DNA glycosylase) protein, in a reaction that requires a covalent intermediate favored under reducing conditions. We set up a new assay based on the capture of plasmid DNA into sensitized microplate wells. DNA damaged by photoactivation of methylene blue was adsorbed on a polylysine-treated plastic well. Then the Fpg protein was added, allowed to fix on the damage by taking advantage of minimized glycosylase activity at low temperature and the reductive trapping of the covalent intermediate, yielding to a stable DNA-protein interaction. The trapped protein was subsequently recognized by a specific antibody. A secondary antibody coupled with horseradish peroxidase was used to detect the complex and the measurement was carried out by chemiluminescence. This new assay offers various potentialities, specifically in the field of technology of ROS producers.


Subject(s)
DNA Damage , Deoxyguanosine/analogs & derivatives , Escherichia coli Proteins , 8-Hydroxy-2'-Deoxyguanosine , DNA Repair , DNA, Bacterial/analysis , DNA, Bacterial/chemistry , DNA, Bacterial/drug effects , DNA-Formamidopyrimidine Glycosylase , Deoxyguanosine/analysis , Escherichia coli/chemistry , Escherichia coli/drug effects , Escherichia coli/metabolism , Evaluation Studies as Topic , Luminescent Measurements , Methods , Methylene Blue , N-Glycosyl Hydrolases/metabolism , Reactive Oxygen Species/metabolism , Sensitivity and Specificity , Substrate Specificity
9.
Biochimie ; 82(1): 25-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10717383

ABSTRACT

The DNA-dependent protein kinase plays a critical role in mammalian DNA double strand break (DSB) repair and in specialized recombination, such as lymphoid V(D)J recombination. Its regulatory subunit Ku (dimer of the Ku70 and Ku80 protein) binds to DNA and recruits the kinase catalytic sub-unit, DNA-PKcs. We show here that three different strains deficient in either the Ku80 (xrs-6) or DNA-PKcs (V-3, scid) component of DNA-PK are markedly sensitive (3.5- to 5-fold) to a group of DNA cross-linking agents, the nitrogen mustards (NMs) (melphalan and mechlorethamine) as compared to their parental cell line. Importantly, the level of hypersensitivity to these drugs was close to the level of hypersensitivity observed for radiomimetic agents that create DSBs in DNA (bleomycin and neocarzinostatin). In addition, sensitivity to NMs was restored to the parental level in the xrs-6 cell line stably transfected with the human Ku80 gene (xrs-6/Ku80), showing unequivocally that DNA-PK is involved in this phenotype. These results indicate that a function of the whole DNA-PK protein complex is involved in the cellular response to NMs and suggest that the repair of DNA interstrand cross-links induced in DNA by NMs involved a DNA-PK dependent pathway that shares common features with DNA DSBs repair.


Subject(s)
Antigens, Nuclear , DNA Helicases , Nitrogen Mustard Compounds/pharmacology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Cell Line/drug effects , Cell Survival/drug effects , Cross-Linking Reagents/pharmacology , DNA/drug effects , DNA Damage/drug effects , DNA-Activated Protein Kinase , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Ku Autoantigen , Mechlorethamine/pharmacology , Melphalan/pharmacology , Mice , Mice, SCID , Mutation , Nuclear Proteins/drug effects , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Transfection , Zinostatin/pharmacology
10.
Mol Cell Biol ; 20(8): 2696-705, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10733572

ABSTRACT

Exposure of mammalian cells to short-wavelength light (UVC) triggers a global response which can either counteract the deleterious effect of DNA damage by enabling DNA repair or lead to apoptosis. Several stress-activated protein kinases participate in this response, making phosphorylation a strong candidate for being involved in regulating the cellular damage response. One factor that is phosphorylated in a UVC-dependent manner is the 32-kDa subunit of the single-stranded DNA-binding replication protein A (RPA32). RPA is required for major cellular processes like DNA replication, and removal of DNA damage by nucleotide excision repair (NER). In this study we examined the signal which triggers RPA32 hyperphosphorylation following UVC irradiation in human cells. Hyperphosphorylation of RPA was observed in cells from patients with either NER or transcription-coupled repair (TCR) deficiency (A, C, and G complementation groups of xeroderma pigmentosum and A and B groups of Cockayne syndrome, respectively). This exclude both NER intermediates and TCR as essential signals for RPA hyperphosphorylation. However, we have observed that UV-sensitive cells deficient in NER and TCR require lower doses of UV irradiation to induce RPA32 hyperphosphorylation than normal cells, indicating that persistent unrepaired lesions contribute to RPA phosphorylation. Finally, the results of UVC irradiation experiments on nonreplicating cells and S-phase-synchronized cells emphasize a major role for DNA replication arrest in the presence of UVC lesions in RPA UVC-induced hyperphosphorylation in mammalian cells.


Subject(s)
DNA Repair , DNA Replication , DNA-Binding Proteins/genetics , Cell Line, Transformed , DNA-Binding Proteins/metabolism , Fibroblasts , Humans , Light , Phosphorylation , Replication Protein A
11.
Food Chem Toxicol ; 37(9-10): 1009-14, 1999.
Article in English | MEDLINE | ID: mdl-10541458

ABSTRACT

Reactive oxygen species (ROS) provoke the formation of base DNA alterations that are processed by an excision step of the lesion followed by a repair synthesis and ligation step to restore the strand continuity. We have reported previously the detection of DNA adducts by an in vitro chemiluminescence DNA repair synthesis assay (Salles et al., 1995) which allows the measurement of repair synthesis by cell-free extracts in damaged plasmid DNA adsorbed on sensitized microplate wells. The 3D (DNA damage detection) assay was performed in the presence of biotin-dUTP which was incorporated during the repair synthesis step. The extent of repair synthesis was measured in an ELISA reaction with ExtrAvidin-horse radish peroxidase and chemiluminescence detection. The 3D assay allows detection of any type of base alterations including base oxidation. Interestingly, under controlled production of ROS a screening procedure of antioxidants might be carried out with the 3D assay. By taking advantage of plasmid DNA adsorption, oxidative base damage can be recognized by the Escherichia coli Fpg protein which was detected in an ELISA reaction with specific antibody and chemiluminescence measurement (4D assay). With the sceening procedure of antioxidative compounds in mind, the development of such assays and their drawbacks are discussed.


Subject(s)
Antioxidants/pharmacology , DNA Repair , Escherichia coli Proteins , DNA Adducts/analysis , DNA Damage , DNA-Formamidopyrimidine Glycosylase , Enzyme-Linked Immunosorbent Assay , Methods , N-Glycosyl Hydrolases , Plasmids , Reactive Oxygen Species/metabolism
13.
Mol Pharmacol ; 56(1): 141-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10385694

ABSTRACT

cis-Dichlorodiammineplatinum(II) (CDDP; cisplatin) is commonly used in combination with ionizing radiation (IR) in the treatment of various malignancies. In vitro, many observations suggest that acquisition of CDDP resistance in cell lines confers cross-resistance to IR, but the molecular mechanisms involved have not been well documented yet. We report here the selection and characterization of a murine CDDP-resistant L1210 cell line (L1210/3R) that exhibits cross-resistance to IR because of an increased capacity to repair double-strand breaks compared with parental cells (L1210/P). In resistant cells, electrophoretic mobility shift assays revealed an increased DNA-end binding activity that could be ascribed, by supershifting the retardation complexes with antibodies, to the autoantigen Ku. The heterodimeric Ku protein, composed of 86-kDa (Ku80) and 70-kDa (Ku70) subunits, is the DNA-targeting component of DNA-dependent protein kinase (DNA-PK), which plays a critical role in mammalian DNA double-strand breaks repair. The increased Ku-binding activity in resistant cells was associated with an overexpression affecting specifically the Ku80 subunit. These data strongly suggest that the increase in Ku activity is responsible for the phenotype of cross-resistance to IR. In addition, these observations, along with previous results from DNA-PK- mutant cells, provide evidence in favor of a role of Ku/DNA-PK in resistance to CDDP. These results suggest that Ku activity may be an important molecular target in cancer therapy at the crossroad between cellular responses to CDDP and IR.


Subject(s)
Antigens, Nuclear , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA Helicases , DNA-Binding Proteins/physiology , Nuclear Proteins/physiology , Animals , DNA Repair , DNA-Activated Protein Kinase , DNA-Binding Proteins/biosynthesis , Dimerization , Drug Resistance, Neoplasm , Kinetics , Ku Autoantigen , Leukemia L1210 , Mice , Nuclear Proteins/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Radiation Tolerance , Radiation, Ionizing , Tumor Cells, Cultured
14.
Biochimie ; 81(1-2): 53-8, 1999.
Article in English | MEDLINE | ID: mdl-10214910

ABSTRACT

The development of in vitro repair assays with human cell-free extracts led to new insights on the mechanism of excision of DNA damage which consists of incision/excision and repair synthesis/ligation. We have adapted the repair synthesis reaction with cells extracts incubated with damaged plasmid DNA performed in liquid phase to solid phase by DNA adsorption into microplate wells. Since cells extracts are repair competent in base excision and nucleotide excision repair, all types of substrate DNA lesions were detected with chemiluminescence measurement after incorporation of biotin-deoxynucleotide during the repair synthesis step. Derivatives of our initial 3D-assay (DNA damage detection) have been set up to: i) screen antioxidative compounds and NER inhibitors; ii) capture genomic DNA (3D(Cell)-assay) that allows detection of alkylated base and consequently determines the kinetics of the cellular repair; and iii) immunodetect the repair proteins in an ELISA reaction (3D(Rec)-assay). The 3D derived assays are presented and discussed.


Subject(s)
DNA Damage , DNA Repair , Animals , Humans , Luminescent Measurements
15.
Biochimie ; 81(1-2): 117-25, 1999.
Article in English | MEDLINE | ID: mdl-10214916

ABSTRACT

The DNA-dependent protein kinase (DNA-PK) is a trimeric nuclear serine/threonine protein kinase consisting of a large catalytic sub-unit and the Ku heterodimer that regulates kinase activity by its association with DNA. DNA-PK is a major component of the DNA double strand break repair apparatus, and cells deficient in one of its component are hypersensitive to ionizing radiation. DNA-PK is also required to lymphoid V(D)J recombination and its absence confers in mice a severe combined immunodeficiency phenotype. The purpose of this review is to summarize the current knowledge on the mechanisms that contribute to regulate DNA-PK activity in vivo or in vitro and relates them to the role of DNA-PK in cellular functions. Finally, the studies devoted to drug-inhibition of DNA-PK in order to enhance cancer therapy by DNA-damaging agents are presented.


Subject(s)
Antigens, Nuclear , DNA Helicases , Eukaryotic Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Eukaryotic Cells/cytology , Ku Autoantigen , Mice , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors
16.
J Biol Chem ; 274(12): 7848-56, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075677

ABSTRACT

The DNA-dependent protein kinase (DNA-PK) is required for double-strand break repair in mammalian cells. DNA-PK contains the heterodimer Ku and a 460-kDa serine/threonine kinase catalytic subunit (p460). Ku binds in vitro to DNA termini or other discontinuities in the DNA helix and is able to enter the DNA molecule by an ATP-independent process. It is clear from in vitro experiments that Ku stimulates the recruitment to DNA of p460 and activates the kinase activity toward DNA-binding protein substrates in the vicinity. Here, we have examined in human nuclear cell extracts the influence of the kinase catalytic activity on Ku binding to DNA. We demonstrate that, although Ku can enter DNA from free ends in the absence of p460 subunit, the kinase activity is required for Ku translocation along the DNA helix when the whole Ku/p460 assembles on DNA termini. When the kinase activity is impaired, DNA-PK including Ku and p460 is blocked at DNA ends and prevents their processing by either DNA polymerization, degradation, or ligation. The control of Ku entry into DNA by DNA-PK catalytic activity potentially represents an important regulation of DNA transactions at DNA termini.


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA Repair , DNA-Binding Proteins/metabolism , DNA/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/pharmacology , Androstadienes/pharmacology , Catalysis , Cell Nucleus/metabolism , DNA-Activated Protein Kinase , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Ku Autoantigen , Wortmannin
17.
Bull Cancer ; 86(12): 977-83, 1999 Dec.
Article in French | MEDLINE | ID: mdl-10660692

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a DNA-activated nuclear serine/threonine protein kinase. DNA-PK consists of a regulatory sub-unit, the heterodimeric Ku protein (composed of a 70- and a 86-kDa subunit) which binds DNA ends and targets the catalytic sub-unit, DNA-PKcs to DNA strand breaks. DNA-PK plays a major role in the repair of double-strand breaks induced in DNA after exposure to ionizing radiation as shown by the extreme radiosensitivity of cells with mutations in Ku86, Ku70 or DNA-PKcs genes. Cells deficient in DNA-PK activity also exhibit hypersensitivity to genotoxic drugs such as cisplatin and nitrogen mustards. In the first part of this review, the current knowledge on the biochemical characteristics of DNA-PK, its mechanism of action in DNA repair and the phenotype of DNA-PK deficient cells is summarized. These results suggest that DNA-PK might play a role in the acquisition of a resistant phenotype of human tumors to radiotherapy, chemotherapy using genotoxic drugs or to both treatments. In the second part of this review, the studies devoted to inhibition of DNA-PK in order to enhance cancer therapy by DNA-damaging agents are presented.


Subject(s)
DNA Damage/radiation effects , DNA Repair , Protein Serine-Threonine Kinases/metabolism , Drug Resistance, Neoplasm , Humans , Phenotype , Protein Serine-Threonine Kinases/genetics , Radiation Tolerance
18.
J Mol Biol ; 284(4): 963-73, 1998 Dec 11.
Article in English | MEDLINE | ID: mdl-9837719

ABSTRACT

We have previously reported that the incision efficiency of the nucleotide excision repair (NER) reaction measured in vitro with cell-free human protein extracts was reduced by up to 80% on a linearized damaged plasmid DNA substrate when compared to supercoiled damaged DNA. The inhibition stemed from the presence of the DNA-end binding Ku70/Ku80 heterodimer which is the regulatory subunit of the DNA-dependent protein kinase (DNA-PK). Here, the origin of the repair inhibition was assessed by a new in vitro assay in which circular or linear plasmid DNA, damaged or undamaged, was quantitatively adsorbed on sensitized microplate wells. The binding of two NER proteins, XPA and p62-TFIIH, indispensable for the incision step of the reaction, was quantified either directly in an ELISA-like reaction in the wells with specific antibodies or in Western blotting experiments on the DNA-bound fraction. We report a dramatic inhibition of XPA and p62-TFIIH association with UVC photoproducts on linear DNA. XPA and p62-TFIIH binding to DNA damage was regained when the reaction was performed with extracts lacking Ku activity (extracts from xrs6 rodent cells) whereas addition of purified human Ku complex to these extracts restored the inhibition. Despite the fact that DNA-PK was active during the NER reaction, the mechanism of inhibition relied on the sole Ku complex, since mutant protein extracts lacking the catalytic DNA-PK subunit (extracts from the human M059J glioma cells) exhibited a strong binding inhibition of XPA and p62-TFIIH proteins on linear damaged DNA, identical to the inhibition observed with the DNA-PK+ control extracts (from M059K cells).


Subject(s)
Antigens, Nuclear , DNA Helicases , DNA Repair , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Transcription Factors, TFII , Animals , CHO Cells , Cell Line , Cricetinae , DNA Damage , DNA, Superhelical/chemistry , DNA, Superhelical/metabolism , DNA-Activated Protein Kinase , DNA-Binding Proteins/chemistry , Dimerization , Humans , Ku Autoantigen , Nuclear Proteins/chemistry , Nucleic Acid Conformation , Plasmids/chemistry , Plasmids/metabolism , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Transcription Factor TFIIH , Transcription Factors/metabolism , Xeroderma Pigmentosum Group A Protein
19.
J Mol Biol ; 281(2): 211-8, 1998 Aug 14.
Article in English | MEDLINE | ID: mdl-9698541

ABSTRACT

We have studied the interactions between DNA damage and human proteins involved in the early steps of nucleotide excision repair (NER) reaction under in vitro conditions with human protein extracts. By using a new assay, we have detected a long-lived DNA/protein complex involving XPA and TFIIH in the course of the NER process. The formation of this complex is exclusively limited to DNA lesions that are substrates of the human excinuclease. We show that, while XPA binding to damaged DNA is ATP-independent, stable association of TFIIH with DNA lesions is promoted by ATP hydrolysis and is dependent on the integrity of XPA and XPC proteins in the cell extract. In addition, XPC is necessary to promote a stable binding of XPA to UV-irradiated DNA. Finally, the co-binding of XPA and TFIIH to DNA damage is correlated to a dose-dependent titration of TFIIH and not XPA from the free protein fraction.


Subject(s)
DNA Repair/physiology , DNA-Binding Proteins/metabolism , DNA/metabolism , Transcription Factors, TFII , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Cell Extracts , Cell-Free System , DNA Damage , Enzyme-Linked Immunosorbent Assay/methods , HeLa Cells , Humans , Hydrolysis , Immunoblotting , Luminescent Measurements , Transcription Factor TFIIH , Ultraviolet Rays , Xeroderma Pigmentosum/genetics , Xeroderma Pigmentosum Group A Protein
20.
Oncogene ; 16(12): 1553-60, 1998 Mar 26.
Article in English | MEDLINE | ID: mdl-9569022

ABSTRACT

The heterodimeric Ku protein, which comprises a 86 kDa (Ku86) amd a 70 kDa (Ku70) subunits, is an abundant nuclear DNA-binding protein which binds in vitro to DNA termini without sequence specificity. Ku is the DNA-targeting component of the large catalytic sub-unit of the DNA-dependent protein kinase complex (DNA-PK[CS]), that plays a critical role in mammalian double-strand break repair and lymphoid V(D)J recombination. By using electrophoretic mobility shift assays, we demonstrated that in addition to the major Ku x DNA complex usually detected in cell line extracts, a second complex with faster electrophoretic mobility was observed in normal peripheral blood lymphocytes (PBL) extracts. The presence of this faster migrating complex was restricted to B cells among the circulating lymphocyte population. Western blot analysis revealed that B cells express a variant form of the Ku86 protein with an apparent molecular weight of 69 kDa, and not the 86 kDa- full-length protein. Although the heterodimer Ku70/variant-Ku86 binds to DNA-ends, this altered form of the Ku heterodimer has a decreased ability to recruit the catalytic component of the complex, DNA-PK(CS), which contributes to an absence of detectable DNA-PK activity in B cells. These data provide a molecular basis for the increased sensitivity of B cells to ionizing radiation and identify a new mechanism of regulation of DNA-PK activity that operates in vivo.


Subject(s)
Antigens, Nuclear , B-Lymphocyte Subsets/enzymology , DNA Helicases , DNA-Binding Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Protein Serine-Threonine Kinases/deficiency , Transcription Factors/biosynthesis , B-Lymphocyte Subsets/metabolism , Cell Line, Transformed , DNA-Activated Protein Kinase , DNA-Binding Proteins/genetics , DNA-Binding Proteins/isolation & purification , Dimerization , Electrophoresis, Polyacrylamide Gel , Genetic Variation , HeLa Cells , Humans , Ku Autoantigen , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Transcription Factors/genetics , Transcription Factors/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...