Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 20(2): 312-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22996684

ABSTRACT

The process of somatic cell reprogramming is gaining increasing interest as reprogrammed cells are considered to hold a great therapeutic potential. However, with current technologies this process is relatively inefficient. Recent studies reported that inhibition of the p53 tumor suppressor profoundly facilitates reprogramming and attributed this effect to the ability of p53 to restrict proliferation and induce apoptosis. Given that mesenchymal-to-epithelial transition (MET) was recently shown to be necessary for reprogramming of fibroblasts, we investigated whether p53 counteracts reprogramming by affecting MET. We found that p53 restricts MET during the early phases of reprogramming and that this effect is primarily mediated by the ability of p53 to inhibit Klf4-dependent activation of epithelial genes. Moreover, transcriptome analysis revealed a large transcriptional signature enriched with epithelial genes, which is markedly induced by Klf4 exclusively in p53(-/-) cells. We also found that the expression of the epithelial marker E-Cadherin negatively correlates with p53 activity in a variety of mesenchymal cells even before the expression of reprogramming factors. Finally, we demonstrate that the inhibitory effect of p53 on MET is mediated by p21. We conclude that inhibition of the p53-p21 axis predisposes mesenchymal cells to the acquisition of epithelial characteristics and renders them more prone to reprogramming. Our study uncovers a novel mechanism by which p53 restrains reprogramming and highlights the role of p53 in regulating cell plasticity.


Subject(s)
Epithelial-Mesenchymal Transition , Tumor Suppressor Protein p53/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Cellular Reprogramming , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
2.
Cell Mol Life Sci ; 66(7): 1209-22, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19099189

ABSTRACT

The FANCJ family of DNA helicases is emerging as an important group of proteins for the prevention of human disease, cancer, and chromosomal instability. FANCJ was identified by its association with breast cancer, and is implicated in Fanconi Anemia. Proteins with sequence similarity to FANCJ are important for maintenance of genomic stability. Mutations in genes encoding proteins related to FANCJ, designated ChlR1 in human and Chl1p in yeast, result in sister chromatid cohesion defects. Nematodes mutated in dog-1 show germline as well as somatic deletions in genes containing guanine-rich DNA. Rtel knockout mice are embryonic lethal, and embryonic stem cells show telomere loss and chromosomal instability. FANCJ also shares sequence similarity with human XPD and yeast RAD3 helicases required for nucleotide excision repair. The recently solved structure of XPD has provided new insight to the helicase core and accessory domains of sequence related Superfamily 2 helicases. The functions and roles of members of the FANCJ-like helicase family will be discussed.


Subject(s)
DNA Helicases/physiology , Fanconi Anemia Complementation Group Proteins/physiology , Genomic Instability/physiology , Amino Acid Sequence , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , DNA Helicases/genetics , DNA Repair/genetics , DNA Repair/physiology , Fanconi Anemia/enzymology , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Genomic Instability/genetics , Humans , Models, Molecular , Molecular Sequence Data , Phylogeny , Protein Conformation , Telomere/genetics , Telomere/physiology
3.
Biochemistry ; 40(50): 15194-202, 2001 Dec 18.
Article in English | MEDLINE | ID: mdl-11735402

ABSTRACT

G-Quadruplex DNAs are folded, non-Watson-Crick structures that can form within guanine-rich DNA sequences such as telomeric repeats. Previous studies have identified a series of trisubstituted acridine derivatives that are potent and selective ligands for G-quadruplex DNA. These ligands have been shown previously to inhibit the activity of telomerase, the specialized reverse transcriptase that regulates telomere length. The RecQ family of DNA helicases, which includes the Bloom's (BLM) and Werner's (WRN) syndrome gene products, are apparently unique among cellular helicases in their ability to efficiently disrupt G-quadruplex DNA. This property may be relevant to telomere maintenance, since it is known that the sole budding yeast RecQ helicase, Sgs1p, is required for a telomerase-independent telomere lengthening pathway reminiscent of the "ALT" pathway in human cells. Here, we show that trisubstituted acridine ligands are potent inhibitors of the helicase activity of the BLM and WRN proteins on both G-quadruplex and B-form DNA substrates. Inhibition of helicase activity is associated with both a reduction in the level of binding of the helicase to G-quadruplex DNA and a reduction in the degree to which the G-quadruplex DNA can support DNA-dependent ATPase activity. We discuss these results in the context of the possible utility of trisubstituted acridines as antitumor agents for the disruption of both telomerase-dependent and telomerase-independent telomere maintenance.


Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , Bloom Syndrome/enzymology , DNA Helicases/antagonists & inhibitors , DNA/pharmacology , Werner Syndrome/enzymology , Acridines/chemistry , Acridines/pharmacology , Adenosine Triphosphatases/genetics , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Base Sequence , Bloom Syndrome/genetics , DNA/chemistry , DNA Helicases/genetics , Humans , In Vitro Techniques , Ligands , Nucleic Acid Conformation , RecQ Helicases , Telomere/drug effects , Werner Syndrome/genetics
4.
Environ Mol Mutagen ; 38(2-3): 227-34, 2001.
Article in English | MEDLINE | ID: mdl-11746759

ABSTRACT

Werner syndrome (WS) is the hallmark premature aging syndrome in which the patients appear much older than their actual chronological age. The disorder is associated with significantly increased genome instability and with transcriptional deficiencies. There has been some uncertainty about whether WS cells are defective in DNA repair. We thus examined repair in vitro in nuclear and mitochondrial DNA. Whereas cellular studies so far do not show significant DNA repair deficiencies, biochemical studies with the Werner protein clearly indicate that it plays a role in DNA repair.


Subject(s)
DNA Repair , Mutagenesis , Werner Syndrome/genetics , Cell Line , DNA Helicases/genetics , DNA, Mitochondrial/genetics , Exodeoxyribonucleases , Humans , RecQ Helicases , Werner Syndrome Helicase
5.
J Biol Chem ; 276(49): 45772-9, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11581270

ABSTRACT

Cockayne Syndrome (CS) is a human genetic disorder with two complementation groups, CS-A and CS-B. The CSB gene product is involved in transcription-coupled repair of DNA damage but may participate in other pathways of DNA metabolism. The present study investigated the role of different conserved helicase motifs of CSB in base excision repair. Stably transformed human cell lines with site-directed CSB mutations in different motifs within its putative helicase domain were established. We find that CSB null and helicase motif V and VI mutants had greater sensitivity than wild type cells to gamma-radiation. Whole cell extracts from CSB null and motif V/VI mutants had lower activity of 8-hydroxyguanine incision in DNA than wild type cells. Also, 8-hydroxyguanine accumulated more in CSB null and motif VI mutant cells than in wild type cells after exposure to gamma-radiation. We conclude that a deficiency in general genome base excision repair of selective modified DNA base(s) might contribute to CS pathogenesis. Furthermore, whereas the disruption of helicase motifs V or VI results in a CSB phenotype, mutations in other helicase motifs do not cause this effect. The biological functions of CSB in different DNA repair pathways may be mediated by distinct functional motifs of the protein.


Subject(s)
Cockayne Syndrome/genetics , DNA Helicases/physiology , DNA Repair/physiology , DNA/genetics , Genome , Guanine/analogs & derivatives , Guanine/chemistry , Amino Acid Sequence , Cell Line, Transformed , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Repair Enzymes , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Oxidative Stress , Poly-ADP-Ribose Binding Proteins
6.
EMBO J ; 20(20): 5791-801, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11598021

ABSTRACT

Werner syndrome (WS) is a human premature aging disorder characterized by chromosomal instability. The cellular defects of WS presumably reflect compromised or aberrant function of a DNA metabolic pathway that under normal circumstances confers stability to the genome. We report a novel interaction of the WRN gene product with the human 5' flap endonuclease/5'-3' exonuclease (FEN-1), a DNA structure-specific nuclease implicated in DNA replication, recombination and repair. WS protein (WRN) dramatically stimulates the rate of FEN-1 cleavage of a 5' flap DNA substrate. The WRN-FEN-1 functional interaction is independent of WRN catalytic function and mediated by a 144 amino acid domain of WRN that shares homology with RecQ DNA helicases. A physical interaction between WRN and FEN-1 is demonstrated by their co-immunoprecipitation from HeLa cell lysate and affinity pull-down experiments using a recombinant C-terminal fragment of WRN. The underlying defect of WS is discussed in light of the evidence for the interaction between WRN and FEN-1.


Subject(s)
DNA Helicases/physiology , Endodeoxyribonucleases/metabolism , Werner Syndrome/genetics , Adenosine Triphosphatases/physiology , Catalysis , DNA/metabolism , DNA Helicases/chemistry , DNA-Binding Proteins/physiology , Endodeoxyribonucleases/chemistry , Enzyme Activation , Exodeoxyribonucleases , Exonucleases/physiology , Flap Endonucleases , HeLa Cells , Humans , Macromolecular Substances , Peptide Fragments/metabolism , Proliferating Cell Nuclear Antigen/physiology , Protein Structure, Tertiary , RecQ Helicases , Recombinant Fusion Proteins/metabolism , Replication Protein A , Werner Syndrome Helicase
7.
J Biol Chem ; 276(48): 44677-87, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11572872

ABSTRACT

Werner syndrome is a human disorder characterized by premature aging, genomic instability, and abnormal telomere metabolism. The Werner syndrome protein (WRN) is the only known member of the RecQ DNA helicase family that contains a 3' --> 5'-exonuclease. However, it is not known whether both activities coordinate in a biological pathway. Here, we describe DNA structures, forked duplexes containing telomeric repeats, that are substrates for the simultaneous action of both WRN activities. We used these substrates to study the interactions between the WRN helicase and exonuclease on a single DNA molecule. WRN helicase unwinds at the forked end of the substrate, whereas the WRN exonuclease acts at the blunt end. Progression of the WRN exonuclease is inhibited by the action of WRN helicase converting duplex DNA to single strand DNA on forks of various duplex lengths. The WRN helicase and exonuclease act in concert to remove a DNA strand from a long forked duplex that is not completely unwound by the helicase. We analyzed the simultaneous action of WRN activities on the long forked duplex in the presence of the WRN protein partners, replication protein A (RPA), and the Ku70/80 heterodimer. RPA stimulated the WRN helicase, whereas Ku stimulated the WRN exonuclease. In the presence of both RPA and Ku, the WRN helicase activity dominated the exonuclease activity.


Subject(s)
Antigens, Nuclear , DNA Helicases/chemistry , DNA Helicases/metabolism , Exodeoxyribonucleases/metabolism , Exodeoxyribonucleases/physiology , Werner Syndrome/metabolism , Base Sequence , Catalysis , DNA/metabolism , DNA-Binding Proteins/metabolism , Dimerization , Dose-Response Relationship, Drug , Exodeoxyribonuclease V , Exonucleases/metabolism , Humans , Kinetics , Ku Autoantigen , Models, Genetic , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Structure, Secondary , RNA/metabolism , RecQ Helicases , Recombinant Proteins/metabolism , Telomere/metabolism , Time Factors , Werner Syndrome Helicase
8.
J Biol Chem ; 276(40): 37076-85, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11477104

ABSTRACT

SecA is an essential ATP-driven motor protein that binds to preproteins and the translocon to promote protein translocation across the eubacterial plasma membrane. Escherichia coli SecA contains seven conserved motifs characteristic of superfamily II of DNA and RNA helicases, and it has been shown previously to possess RNA helicase activity. SecA has also been shown to be an autogenous repressor that binds to its translation initiation region on secM-secA mRNA, thereby blocking and dissociating 30 S ribosomal subunits. Here we show that SecA is an ATP-dependent helicase that unwinds a mimic of the repressor helix of secM-secA mRNA. Mutational analysis of the seven conserved helicase motifs in SecA allowed us to identify mutants that uncouple SecA-dependent protein translocation activity from its helicase activity. Helicase-defective secA mutants displayed normal protein translocation activity and autogenous repression of secA in vivo. Our studies indicate that SecA helicase activity is nonessential and does not appear to be necessary for efficient protein secretion and secA autoregulation.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , DNA Helicases/metabolism , Escherichia coli Proteins , Escherichia coli/enzymology , Membrane Transport Proteins/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Translocation/physiology , Conserved Sequence , DNA Helicases/genetics , DNA, Bacterial/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Homeostasis , Membrane Transport Proteins/genetics , Molecular Sequence Data , Mutation , Nucleic Acid Conformation , Protein Transport , RNA, Bacterial/metabolism , SEC Translocation Channels , SecA Proteins , Sequence Homology, Amino Acid
9.
Nucleic Acids Res ; 29(13): 2843-9, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11433031

ABSTRACT

BLM and WRN, the products of the Bloom's and Werner's syndrome genes, are members of the RecQ family of DNA helicases. Although both have been shown previously to unwind simple, partial duplex DNA substrates with 3'-->5' polarity, little is known about the structural features of DNA that determine the substrate specificities of these enzymes. We have compared the substrate specificities of the BLM and WRN proteins using a variety of partial duplex DNA molecules, which are based upon a common core nucleotide sequence. We show that neither BLM nor WRN is capable of unwinding duplex DNA from a blunt-ended terminus or from an internal nick. However, both enzymes efficiently unwind the same blunt-ended duplex containing a centrally located 12 nt single-stranded 'bubble', as well as a synthetic X-structure (a model for the Holliday junction recombination intermediate) in which each 'arm' of the 4-way junction is blunt-ended. Surprisingly, a 3'-tailed duplex, a standard substrate for 3'-->5' helicases, is unwound much less efficiently by BLM and WRN than are the bubble and X-structure substrates. These data show conclusively that a single-stranded 3'-tail is not a structural requirement for unwinding of standard B-form DNA by these helicases. BLM and WRN also both unwind a variety of different forms of G-quadruplex DNA, a structure that can form at guanine-rich sequences present at several genomic loci. Our data indicate that BLM and WRN are atypical helicases that are highly DNA structure specific and have similar substrate specificities. We interpret these data in the light of the genomic instability and hyper-recombination characteristics of cells from individuals with Bloom's or Werner's syndrome.


Subject(s)
Bloom Syndrome/enzymology , DNA Helicases/metabolism , DNA/chemistry , DNA/metabolism , Nucleic Acid Conformation , Werner Syndrome/enzymology , Base Sequence , Bloom Syndrome/genetics , Crossing Over, Genetic/genetics , DNA/genetics , DNA Helicases/genetics , Humans , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , Substrate Specificity , Werner Syndrome/genetics
10.
J Biol Chem ; 276(37): 35093-102, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11427532

ABSTRACT

Werner syndrome (WS) is characterized by the early onset of symptoms of premature aging, cancer, and genomic instability. The molecular basis of the defects is not understood but presumably relates to the DNA helicase and exonuclease activities of the protein encoded by the WRN gene that is mutated in the disease. The attenuation of p53-mediated apoptosis in WS cells and reported physical interaction between WRN and the tumor suppressor p53 suggest that p53 and WRN functionally interact in a pathway necessary for the normal cellular response. In this study, we have demonstrated that p53 inhibits the exonuclease activity of the purified full-length recombinant WRN protein. p53 did not have an effect on a truncated amino-terminal WRN fragment that retains exonuclease activity but lacks the physical interaction domain for p53 located in the carboxyl terminus. Two naturally occurring p53 mutants found in human cancer displayed a reduced ability to inhibit WRN exonuclease activity. In cells arrested in S phase with hydroxyurea, WRN exits the nucleolus and colocalizes with p53 in the nucleoplasm. The regulation of WRN function by p53 is likely to play an important role in the maintenance of genomic integrity and prevention of cancer and other clinical symptoms associated with WS.


Subject(s)
DNA Helicases/physiology , Exodeoxyribonucleases/antagonists & inhibitors , Tumor Suppressor Protein p53/physiology , Werner Syndrome/genetics , Adenosine Triphosphatases/metabolism , Apoptosis , Base Sequence , Catalysis , DNA Damage , DNA Helicases/analysis , DNA Helicases/chemistry , DNA Helicases/metabolism , Humans , Molecular Sequence Data , RecQ Helicases , Tumor Suppressor Protein p53/analysis , Tumor Suppressor Protein p53/chemistry , Werner Syndrome Helicase
11.
J Biol Chem ; 276(5): 3024-30, 2001 Feb 02.
Article in English | MEDLINE | ID: mdl-11110789

ABSTRACT

Bloom syndrome and Werner syndrome are genome instability disorders, which result from mutations in two different genes encoding helicases. Both enzymes are members of the RecQ family of helicases, have a 3' --> 5' polarity, and require a 3' single strand tail. In addition to their activity in unwinding duplex substrates, recent studies show that the two enzymes are able to unwind G2 and G4 tetraplexes, prompting speculation that failure to resolve these structures in Bloom syndrome and Werner syndrome cells may contribute to genome instability. The triple helix is another alternate DNA structure that can be formed by sequences that are widely distributed throughout the human genome. Here we show that purified Bloom and Werner helicases can unwind a DNA triple helix. The reactions are dependent on nucleoside triphosphate hydrolysis and require a free 3' tail attached to the third strand. The two enzymes unwound triplexes without requirement for a duplex extension that would form a fork at the junction of the tail and the triplex. In contrast, a duplex formed by the third strand and a complement to the triplex region was a poor substrate for both enzymes. However, the same duplex was readily unwound when a noncomplementary 5' tail was added to form a forked structure. It seems likely that structural features of the triplex mimic those of a fork and thus support efficient unwinding by the two helicases.


Subject(s)
Bloom Syndrome/enzymology , DNA Helicases/metabolism , DNA/metabolism , Nucleic Acid Denaturation , Werner Syndrome/enzymology , Binding Sites , Humans , Nucleic Acid Conformation
12.
Exp Gerontol ; 35(6-7): 695-702, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11053659

ABSTRACT

Werner syndrome is a premature aging syndrome displaying numerous signs and symptoms found in normal aging. The disease is associated with a mutation in the WRN gene. We have purified the Werner protein (WRN) and studied its biochemical activities and its protein interactions. WRN is a helicase and an exonuclease and also has an associated ATPase activity. WRN interacts physically and functionally with replication protein A (RPA), which stimulates its helicase activity. We have studied the WRN exonuclease activity and found that it can be blocked by certain DNA lesions and not by others. Thus, while WRN does not bind to DNA damage, it may have properties that allow it to sense the presence of damage in DNA. More recently we have found other protein interactions that involve physical and functional interactions with WRN, which could suggest a role for WRN in DNA repair.


Subject(s)
Antigens, Nuclear , DNA Helicases/physiology , Werner Syndrome/etiology , DNA Damage , DNA Helicases/genetics , DNA Repair , DNA-Binding Proteins/physiology , Exodeoxyribonucleases , Humans , Ku Autoantigen , Nuclear Proteins/physiology , RecQ Helicases , Werner Syndrome/genetics , Werner Syndrome Helicase
13.
Nucleic Acids Res ; 28(16): 3151-9, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10931931

ABSTRACT

Cockayne syndrome (CS) is a human genetic disorder characterized by post-natal growth failure, neurological abnormalities and premature aging. CS cells exhibit high sensitivity to UV light, delayed RNA synthesis recovery after UV irradiation and defective transcription-coupled repair (TCR). Two genetic complementation groups of CS have been identified, designated CS-A and CS-B. The CSB gene encodes a helicase domain and a highly acidic region N-terminal to the helicase domain. This study describes the genetic characterization of a CSB mutant allele encoding a full deletion of the acidic region. We have tested its ability to complement the sensitivity of UV61, the hamster homolog of human CS-B cells, to UV and the genotoxic agent N-acetoxy-2-acetylaminofluorene (NA-AAF). Deleting 39 consecutive amino acids, of which approximately 60% are negatively charged, did not impact on the ability of the protein to complement the sensitive phenotype of UV61 cells to either UV or NA-AAF. Our data indicate that the highly acidic region of CSB is not essential for the TCR and general genome repair pathways of UV- and NA-AAF-induced DNA lesions.


Subject(s)
Apoptosis , Cockayne Syndrome/genetics , DNA Helicases/genetics , DNA Repair , Sequence Deletion , Acetoxyacetylaminofluorene/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Cricetinae , DNA Helicases/metabolism , DNA Repair/drug effects , DNA Repair/radiation effects , DNA Repair Enzymes , Genetic Complementation Test , Humans , Molecular Sequence Data , Poly-ADP-Ribose Binding Proteins , Proliferating Cell Nuclear Antigen/metabolism , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Ultraviolet Rays
14.
Nucleic Acids Res ; 28(12): 2420-30, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10871376

ABSTRACT

Maintenance of genomic integrity is vital to all organisms. A number of human genetic disorders, including Werner Syndrome, Bloom Syndrome and Rothmund-Thomson Syndrome, exhibit genomic instability with some phenotypic characteristics of premature aging and cancer predisposition. Presumably the aberrant cellular and clinical phenotypes in these disorders arise from defects in important DNA metabolic pathways such as replication, recombination or repair. These syndromes are all characterized by defects in a member of the RecQ family of DNA helicases. To obtain a better understanding of how these enzymes function in DNA metabolic pathways that directly influence chromosomal integrity, we have examined the effects of non-covalent DNA modifications on the catalytic activities of purified Werner (WRN) and Bloom (BLM) DNA helicases. A panel of DNA-binding ligands displaying unique properties for interacting with double helical DNA was tested for their effects on the unwinding activity of WRN and BLM helicases on a partial duplex DNA substrate. The levels of inhibition by a number of these compounds were distinct from previously reported values for viral, prokaryotic and eukaryotic helicases. The results demonstrate that BLM and WRN proteins exhibit similar sensitivity profiles to these DNA-binding ligands and are most potently inhibited by the structurally related minor groove binders distamycin A and netropsin (K(i)

Subject(s)
Adenosine Triphosphatases/antagonists & inhibitors , DNA Helicases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Intercalating Agents/pharmacology , Adenosine Triphosphatases/chemistry , Bloom Syndrome/enzymology , DNA Helicases/chemistry , Distamycins/pharmacology , Enzyme Inhibitors/chemistry , Exodeoxyribonucleases , Humans , Intercalating Agents/chemistry , Kinetics , Ligands , Netropsin/pharmacology , RecQ Helicases , Recombinant Proteins/antagonists & inhibitors , Topoisomerase I Inhibitors , Werner Syndrome/enzymology , Werner Syndrome Helicase
15.
J Biol Chem ; 275(31): 23500-8, 2000 Aug 04.
Article in English | MEDLINE | ID: mdl-10825162

ABSTRACT

Bloom's syndrome is a rare autosomal recessive disorder characterized by genomic instability and predisposition to cancer. BLM, the gene defective in Bloom's syndrome, encodes a 159-kDa protein possessing DNA-stimulated ATPase and ATP-dependent DNA helicase activities. We have examined mechanistic aspects of the catalytic functions of purified recombinant BLM protein. Through analyzing the effects of different lengths of DNA cofactor on ATPase activity, we provide evidence to suggest that BLM translocates along single-stranded DNA in a processive manner. The helicase reaction catalyzed by BLM protein was examined as a function of duplex DNA length. We show that BLM catalyzes unwinding of short DNA duplexes (/=259-bp). The presence of the human single-stranded DNA-binding protein (human replication protein A (hRPA)) stimulates the BLM unwinding reaction on the 259-bp partial duplex DNA substrate. Heterologous single-stranded DNA-binding proteins fail to stimulate similarly the helicase activity of BLM protein. This is the first demonstration of a functional interaction between BLM and another protein. Consistent with a functional interaction between hRPA and the BLM helicase, we demonstrate a direct physical interaction between the two proteins mediated by the 70-kDa subunit of RPA. The interactions between BLM and hRPA suggest that the two proteins function together in vivo to unwind DNA duplexes during replication, recombination, or repair.


Subject(s)
Adenosine Triphosphatases/metabolism , Bloom Syndrome , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Blotting, Western , DNA/metabolism , DNA Helicases/genetics , DNA Replication , Enzyme-Linked Immunosorbent Assay , Humans , Hydrolysis , Movement , Nucleic Acid Conformation , Protein Binding , RecQ Helicases , Recombinant Proteins/metabolism , Recombination, Genetic , Replication Protein A , Saccharomyces cerevisiae , Species Specificity
16.
Genes Dev ; 14(8): 907-12, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10783163

ABSTRACT

Werner syndrome (WS) is the hallmark premature aging disorder in which affected humans appear older than their chronological age. The protein WRNp, defective in WS, has helicase function, DNA-dependent ATPase, and exonuclease activity. Although WRNp functions in nucleic acid metabolism, there is little or no information about the pathways or protein interactions in which it participates. Here we identify Ku70 and Ku86 as proteins that interact with WRNp. Although Ku proteins had no effect on ATPase or helicase activity, they strongly stimulated specific exonuclease activity. These results suggest that WRNp and the Ku complex participate in a common DNA metabolic pathway.


Subject(s)
Antigens, Nuclear , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Adenosine Triphosphatases/metabolism , Animals , Base Sequence , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Chromatography, Affinity , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Exodeoxyribonucleases , Exonucleases/metabolism , Humans , Ku Autoantigen , Molecular Sequence Data , Nuclear Proteins/chemistry , Precipitin Tests , Protein Binding , RecQ Helicases , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Werner Syndrome Helicase
17.
Oncogene ; 19(4): 477-89, 2000 Jan 27.
Article in English | MEDLINE | ID: mdl-10698517

ABSTRACT

Cockayne syndrome (CS) is a human autosomal recessive disorder characterized by many neurological and developmental abnormalities. CS cells are defective in the transcription coupled repair (TCR) pathway that removes DNA damage from the transcribed strand of active genes. The individuals suffering from CS do not generally develop cancer but show increased neurodegeneration. Two genetic complementation groups (CS-A and CS-B) have been identified. The lack of cancer formation in CS may be due to selective elimination of cells containing DNA damage by a suicidal pathway. In this study, we have evaluated the role of the CSB gene in UV induced apoptosis in human and hamster cells. The hamster cell line UV61 carries a mutation in the homolog of the human CSB gene. We show that both human CS-B and hamster UV61 cells display increased apoptotic response following UV exposure compared with normal cells. The increased sensitivity of UV61 cells to apoptosis is complemented by the transfection of the wild type human CSB gene. In order to determine which functional domain of the CSB gene participates in the apoptotic pathway, we constructed stable cell lines with different CSB domain disruptions. UV61 cells were stably transfected with the human CSB cDNA containing a point mutation in the highly conserved glutamic acid residue in ATPase motif II. This cell line (UV61/ pc3.1-CSBE646Q) showed the same increased apoptosis as the UV61 cells. In contrast, cells containing a deletion in the acidic domain at the N-terminal end of the CSB protein had no effect on apoptosis. This indicates that the integrity of the ATPase domain of CSB protein is critical for preventing the UV induced apoptotic pathway. In primary human CS-B cells, the induction and stabilization of the p53 protein seems to correlate with their increased apoptotic potential. In contrast, no change in the level of either p53 or activation of mdm2 protein by p53 was observed in hamster UV61 cells after UV exposure. This suggests that the CSB dependent apoptotic pathway can occur independently of the transactivation potential of p53 in hamster cells.


Subject(s)
Adenosine Triphosphatases/physiology , Apoptosis/radiation effects , Cockayne Syndrome/pathology , DNA Helicases/physiology , DNA Repair/genetics , Nuclear Proteins , Proto-Oncogene Proteins c-bcl-2 , Ultraviolet Rays , Amino Acid Sequence , Animals , Apoptosis/physiology , Cell Line , Cockayne Syndrome/enzymology , Cockayne Syndrome/genetics , Cricetinae , Cricetulus , DNA/biosynthesis , DNA Helicases/chemistry , DNA Repair Enzymes , Genes, p53 , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Point Mutation , Poly-ADP-Ribose Binding Proteins , Protein Structure, Tertiary , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-mdm2 , Proto-Oncogene Proteins p21(ras)/analysis , RNA/biosynthesis , RNA Polymerase II/antagonists & inhibitors , Radiation Tolerance/genetics , Recombinant Fusion Proteins/physiology , Sequence Deletion , Transcriptional Activation , Transfection , Tumor Suppressor Protein p53/physiology , Ultraviolet Rays/adverse effects , bcl-2-Associated X Protein
18.
Electrophoresis ; 21(3): 509-16, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10726750

ABSTRACT

A procedure was established for selecting phage antibodies (phage-abs) from phage-displayed antibody repertoires by panning against proteins, separated by sodium dodecyl phosphate-polyacrylamide gel electrophoresis (SDS-PAGE) and electroblotted onto nitrocellulose membranes (Western blots). This immobilization strategy is applicable for secondary rounds of panning in selections against semipurified proteins, and directs the selection toward antibodies suitable as immunochemical reagents in Western blots. In model experiments, enrichment factors as high as 1.9x10(5) were obtained in a single round of panning. Furthermore, we demonstrate the application of this approach by selection of phage-abs recognizing the human Werner protein, which is defective in a premature aging syndrome.


Subject(s)
Antibodies, Viral/immunology , Bacteriophages/immunology , DNA Helicases/immunology , Base Sequence , Blotting, Western , DNA Fingerprinting , DNA Primers , Enzyme-Linked Immunosorbent Assay , Exodeoxyribonucleases , Humans , Polymerase Chain Reaction , RecQ Helicases , Werner Syndrome Helicase
19.
EMBO Rep ; 1(1): 80-4, 2000 Jul.
Article in English | MEDLINE | ID: mdl-11256630

ABSTRACT

Individuals affected by the autosomal recessive disorder Werner's syndrome (WS) develop many of the symptoms characteristic of premature ageing. Primary fibroblasts cultured from WS patients exhibit karyotypic abnormalities and a reduced replicative life span. The WRN gene encodes a 3'-5' DNA helicase, and is a member of the RecQ family, which also includes the product of the Bloom's syndrome gene (BLM). In this work, we show that WRN promotes the ATP-dependent translocation of Holliday junctions, an activity that is also exhibited by BLM. In cells arrested in S-phase with hydroxyurea, WRN localizes to discrete nuclear foci that coincide with those formed by the single-stranded DNA binding protein replication protein A. These results are consistent with a model in which WRN prevents aberrant recombination events at sites of stalled replication forks by dissociating recombination intermediates.


Subject(s)
DNA Helicases/metabolism , DNA Replication , DNA/metabolism , Recombination, Genetic , Werner Syndrome/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Nucleus/metabolism , DNA/genetics , DNA Helicases/genetics , Exodeoxyribonucleases , HeLa Cells , Humans , Microscopy, Fluorescence , RecQ Helicases , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Werner Syndrome/genetics , Werner Syndrome Helicase
20.
Mol Biol Cell ; 10(11): 3583-94, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10564257

ABSTRACT

Cockayne syndrome (CS) is a human genetic disorder characterized by UV sensitivity, developmental abnormalities, and premature aging. Two of the genes involved, CSA and CSB, are required for transcription-coupled repair (TCR), a subpathway of nucleotide excision repair that removes certain lesions rapidly and efficiently from the transcribed strand of active genes. CS proteins have also been implicated in the recovery of transcription after certain types of DNA damage such as those lesions induced by UV light. In this study, site-directed mutations have been introduced to the human CSB gene to investigate the functional significance of the conserved ATPase domain and of a highly acidic region of the protein. The CSB mutant alleles were tested for genetic complementation of UV-sensitive phenotypes in the human CS-B homologue of hamster UV61. In addition, the CSB mutant alleles were tested for their ability to complement the sensitivity of UV61 cells to the carcinogen 4-nitroquinoline-1-oxide (4-NQO), which introduces bulky DNA adducts repaired by global genome repair. Point mutation of a highly conserved glutamic acid residue in ATPase motif II abolished the ability of CSB protein to complement the UV-sensitive phenotypes of survival, RNA synthesis recovery, and gene-specific repair. These data indicate that the integrity of the ATPase domain is critical for CSB function in vivo. Likewise, the CSB ATPase point mutant failed to confer cellular resistance to 4-NQO, suggesting that ATP hydrolysis is required for CSB function in a TCR-independent pathway. On the contrary, a large deletion of the acidic region of CSB protein did not impair the genetic function in the processing of either UV- or 4-NQO-induced DNA damage. Thus the acidic region of CSB is likely to be dispensable for DNA repair, whereas the ATPase domain is essential for CSB function in both TCR-dependent and -independent pathways.


Subject(s)
Adenosine Triphosphatases/genetics , DNA Helicases/genetics , DNA Repair/genetics , 4-Nitroquinoline-1-oxide/pharmacology , Adenosine Triphosphatases/chemistry , Amino Acid Sequence , Animals , Cell Line , Cell Survival , Clone Cells/radiation effects , Cockayne Syndrome/genetics , Cricetinae , DNA Damage , DNA Helicases/chemistry , DNA Repair Enzymes , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Poly-ADP-Ribose Binding Proteins , Pyrimidine Dimers/genetics , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Transfection , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...