Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters










Publication year range
1.
Oncogene ; 35(37): 4836-45, 2016 09 15.
Article in English | MEDLINE | ID: mdl-26876210

ABSTRACT

Rad54 and Mus81 mammalian proteins physically interact and are important for the homologous recombination DNA repair pathway; however, their functional interactions in vivo are poorly defined. Here, we show that combinatorial loss of Rad54 and Mus81 results in hypersensitivity to DNA-damaging agents, defects on both the homologous recombination and non-homologous DNA end joining repair pathways and reduced fertility. We also observed that while Mus81 deficiency diminished the cleavage of common fragile sites, very strikingly, Rad54 loss impaired this cleavage to even a greater extent. The inefficient repair of DNA double-strand breaks (DSBs) in Rad54(-/-)Mus81(-/-) cells was accompanied by elevated levels of chromosome missegregation and cell death. Perhaps as a consequence, tumor incidence in Rad54(-/-)Mus81(-/-) mice remained comparable to that in Mus81(-/-) mice. Our study highlights the importance of the cooperation between Rad54 and Mus81 for mediating DNA DSB repair and restraining chromosome missegregation.


Subject(s)
DNA Helicases/genetics , DNA Repair/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Neoplasms/genetics , Nuclear Proteins/genetics , Animals , Chromosomes/genetics , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair/genetics , Homologous Recombination/genetics , Humans , Mice , Mice, Knockout , Neoplasms/pathology
2.
Br J Cancer ; 104(4): 653-63, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21266972

ABSTRACT

AIMS: Modulation of DNA base excision repair (BER) has the potential to enhance response to chemotherapy and improve outcomes in tumours such as melanoma and glioma. APE1, a critical protein in BER that processes potentially cytotoxic abasic sites (AP sites), is a promising new target in cancer. In the current study, we aimed to develop small molecule inhibitors of APE1 for cancer therapy. METHODS: An industry-standard high throughput virtual screening strategy was adopted. The Sybyl8.0 (Tripos, St Louis, MO, USA) molecular modelling software suite was used to build inhibitor templates. Similarity searching strategies were then applied using ROCS 2.3 (Open Eye Scientific, Santa Fe, NM, USA) to extract pharmacophorically related subsets of compounds from a chemically diverse database of 2.6 million compounds. The compounds in these subsets were subjected to docking against the active site of the APE1 model, using the genetic algorithm-based programme GOLD2.7 (CCDC, Cambridge, UK). Predicted ligand poses were ranked on the basis of several scoring functions. The top virtual hits with promising pharmaceutical properties underwent detailed in vitro analyses using fluorescence-based APE1 cleavage assays and counter screened using endonuclease IV cleavage assays, fluorescence quenching assays and radiolabelled oligonucleotide assays. Biochemical APE1 inhibitors were then subjected to detailed cytotoxicity analyses. RESULTS: Several specific APE1 inhibitors were isolated by this approach. The IC(50) for APE1 inhibition ranged between 30 nM and 50 µM. We demonstrated that APE1 inhibitors lead to accumulation of AP sites in genomic DNA and potentiated the cytotoxicity of alkylating agents in melanoma and glioma cell lines. CONCLUSIONS: Our study provides evidence that APE1 is an emerging drug target and could have therapeutic application in patients with melanoma and glioma.


Subject(s)
Brain Neoplasms/drug therapy , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/therapeutic use , Glioma/drug therapy , Melanoma/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , Drug Discovery , Drug Evaluation, Preclinical , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Glioma/pathology , HeLa Cells , High-Throughput Screening Assays/methods , Humans , Inhibitory Concentration 50 , Melanoma/pathology , Models, Biological , Models, Molecular , Structure-Activity Relationship
3.
Oncogene ; 29(33): 4705-14, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20531307

ABSTRACT

BLM is a RecQ family helicase that is defective in individuals with the cancer predisposition disorder, Bloom's syndrome (BS). At the cellular level, BS is characterized by hyper-recombination manifested as excessive sister chromatid exchange and loss of heterozygosity. However, the precise function of BLM remains unclear. Multiple roles have been proposed for BLM in the homologous recombination (HR) repair pathway, including 'early' functions, such as the stimulation of resection of DNA double-strand break ends or displacement of the invading strand of DNA displacement loops, and 'late' roles, such as dissolution of double Holliday junctions. However, most of the evidence for these putative roles comes from in vitro biochemical data. In this study, we report the characterization of mouse embryonic stem cells with disruption of Blm and/or Rad54 genes. We show that Blm has roles both upstream and downstream of the Rad54 protein, a core HR factor. Disruption of Rad54 in the Blm-mutant background reduced the elevated level of gene targeting and of sister chromatid exchanges, implying that Blm primarily functions downstream of Rad54 in the HR pathway. Conversely, however, mutation of Blm in Rad54(-/-) cells rescued their mitomycin C (MMC) sensitivity, and decreased both the level of DNA damage and cell cycle perturbation induced by MMC, suggesting an early role for Blm. Our data are consistent with Blm having at least two roles in HR repair in mammalian cells.


Subject(s)
DNA Repair , Embryonic Stem Cells/physiology , RecQ Helicases/genetics , Recombination, Genetic , Animals , Cell Line, Tumor , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Replication , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Gene Targeting , Immunoblotting , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RecQ Helicases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sister Chromatid Exchange
4.
Cytogenet Genome Res ; 125(3): 165-75, 2009.
Article in English | MEDLINE | ID: mdl-19738377

ABSTRACT

The Bloom protein (BLM) and Topoisomerase IIIalpha are found in association with proteins of the Fanconi anemia (FA) pathway, a disorder manifesting increased cellular sensitivity to DNA crosslinking agents. In order to determine if the association reflects a functional interaction for the maintenance of genome stability, we have analyzed the effects of siRNA-mediated depletion of the proteins in human cells. Depletion of Topoisomerase IIIalpha or BLM leads to increased radial formation, as is seen in FA. BLM and Topoisomerase IIIalpha are epistatic to the FA pathway for suppression of radial formation in response to DNA interstrand crosslinks since depletion of either of them in FA cells does not increase radial formation. Depletion of Topoisomerase IIIalpha or BLM also causes an increase in sister chromatid exchanges, as is seen in Bloom syndrome cells. Human Fanconi anemia cells, however, do not demonstrate increased sister chromatid exchanges, separating this response from radial formation. Primary cell lines from mice defective in both Blm and Fancd2 have the same interstrand crosslink-induced genome instability as cells from mice deficient in the Fancd2 protein alone. These observations demonstrate that the association of BLM and Topoisomerase IIIalpha with Fanconi proteins is a functional one, delineating a BLM-Topoisomerase IIIalpha-Fanconi pathway that is critical for suppression of chromosome radial formation.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA/metabolism , Fanconi Anemia/metabolism , RecQ Helicases/metabolism , Animals , Cell Line , Cross-Linking Reagents/pharmacology , DNA Topoisomerases, Type I/genetics , Fanconi Anemia/genetics , Genomic Instability/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitomycin/pharmacology , RNA, Small Interfering/genetics , RecQ Helicases/genetics , Sister Chromatid Exchange
5.
Cell Mol Life Sci ; 64(17): 2306-22, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17571213

ABSTRACT

The RecQ helicases belong to the Superfamily II group of DNA helicases, and are defined by amino acid motifs that show sequence similarity to the catalytic domain of Escherichia coli RecQ. RecQ helicases have crucial roles in the maintenance of genome stability. In humans, there are five RecQ helicases and deficiencies in three of them cause genetic disorders characterised by cancer predisposition, premature aging and/or developmental abnormalities. RecQ helicase-deficient cells exhibit aberrant genetic recombination and/or DNA replication, which result in chromosomal instability and a decreased potential for proliferation. Here, we review the current knowledge of the molecular genetics of RecQ helicases, focusing on the human RecQ helicase disorders and mouse models of these conditions.


Subject(s)
DNA Repair-Deficiency Disorders/genetics , RecQ Helicases/physiology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Animals , Bloom Syndrome/diagnosis , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , Cellular Senescence/genetics , DNA Damage , DNA Helicases/genetics , DNA Helicases/physiology , DNA Repair-Deficiency Disorders/diagnosis , DNA Repair-Deficiency Disorders/metabolism , Disease Models, Animal , Humans , Mice , Phenotype , RecQ Helicases/chemistry , RecQ Helicases/genetics , Recombination, Genetic , Syndrome , Werner Syndrome/diagnosis , Werner Syndrome/genetics , Werner Syndrome/metabolism
6.
Oncogene ; 25(14): 2119-23, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16288211

ABSTRACT

A subset of DNA helicases, the RecQ family, has been found to be associated with the p53-mediated apoptotic pathway and is involved in maintaining genomic integrity. This family contains the BLM and WRN helicases, in which germline mutations are responsible for Bloom and Werner syndromes, respectively. TFIIH DNA helicases, XPB and XPD, are also components in this apoptotic pathway. We hypothesized that there may be some redundancy between helicases in their ability to complement the attenuated p53-mediated apoptotic levels seen in cells from individuals with diseases associated with these defective helicase genes. The attenuated apoptotic phenotype in Bloom syndrome cells was rescued not only by ectopic expression of BLM, but also by WRN or XPB, both 3' --> 5' helicases, but not expression of the 5' --> 3' helicase XPD. Overexpression of Sgs1, a WRN/BLM yeast homolog, corrected the reduction in BS cells only, which is consistent with Sgs1 being evolutionarily most homologous to BLM. A restoration of apoptotic levels in cells from WS, XPB or XPD patients was attained only by overexpression of the specific helicase. Our data suggest a limited redundancy in the pathways of these RecQ helicases in p53-induced apoptosis.


Subject(s)
Apoptosis/physiology , DNA Helicases/metabolism , Tumor Suppressor Protein p53/physiology , Bloom Syndrome/enzymology , Germ-Line Mutation , Humans , Werner Syndrome/enzymology
7.
Biochem Soc Trans ; 33(Pt 6): 1456-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16246145

ABSTRACT

The RecQ family of DNA helicases is highly conserved in evolution from bacteria to humans. Of the five known human RecQ family members, three (BLM, WRN and RECQ4, which cause Bloom's syndrome, Werner's syndrome and Rothmund-Thomson syndrome respectively) are mutated in distinct clinical disorders associated with cancer predisposition and/or premature aging. BLM forms part of a multienzyme complex including topoisomerase IIIalpha, replication protein A and a newly identified factor called BLAP75. Together, these proteins play a role in the resolution of DNA structures that arise during the process of homologous recombination repair. In the absence of BLM, cells show genomic instability and a high incidence of sister-chromatid exchanges. In addition to a DNA structure-specific helicase activity, BLM also catalyses Holliday-junction branch migration and the annealing of complementary single-stranded DNA molecules.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Genomic Instability , Recombination, Genetic , Bloom Syndrome/genetics , Bloom Syndrome/metabolism , DNA, Cruciform , DNA, Single-Stranded , Exodeoxyribonucleases , Humans , Nucleic Acid Conformation , RecQ Helicases , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase
8.
Br J Cancer ; 93(1): 60-9, 2005 Jul 11.
Article in English | MEDLINE | ID: mdl-15956976

ABSTRACT

A pharmacokinetically guided phase I study of topotecan and etoposide phosphate was conducted in recurrent ovarian cancer. The scheduling of the topoisomerase I and II inhibitors was determined using in vitro activity data. All patients had recurrent disease following prior platinum-containing chemotherapy. Patients had a World Health Organisation performance status of 0-2 and adequate bone marrow, renal and hepatic function. Treatment was with topotecan intravenously for 5 days followed immediately by a 5-day intravenous infusion of etoposide phosphate (EP), with pharmacokinetically guided dose adjustment. Plasma etoposide levels were measured on days 2 and 4 of the infusion. A total of 21 patients entered the study. In all, 48% were platinum resistant and 71% had received prior paclitaxel. The main toxicities were haematological, short lived and reversible. A total of 29% of patients experienced grade 4 thrombocytopenia and 66% grade 4 neutropenia after the first cycle. Neutropenia and thrombocytopenia was dose limiting. The maximum-tolerated dose was topotecan 0.85 mg m(-2) day(-1) days 1-5 followed immediately by a 5-day infusion of EP at a plasma concentration of 1 mug ml(-1). The response rate (RR) was 28% in 18 evaluable patients. There was marked interpatient variability in topoisomerase IIalpha levels measured from peripheral lymphocytes, with no observed increase following topotecan. This regimen of topotecan followed by EP demonstrated good activity in recurrent ovarian cancer and was noncrossresistant with paclitaxel. Both the toxicity and RR was higher than would be expected from the single agent data, in keeping with synergy of action.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Ovarian Neoplasms/drug therapy , Adult , Aged , Antigens, Neoplasm/blood , Antineoplastic Combined Chemotherapy Protocols/adverse effects , DNA Topoisomerases, Type II/blood , DNA-Binding Proteins/blood , Etoposide/administration & dosage , Etoposide/adverse effects , Etoposide/analogs & derivatives , Etoposide/pharmacokinetics , Female , Humans , Middle Aged , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/adverse effects , Organophosphorus Compounds/pharmacokinetics , Quality of Life , Recurrence , Topotecan/administration & dosage , Topotecan/adverse effects , Topotecan/pharmacokinetics
9.
Biochem Soc Trans ; 32(Pt 6): 957-8, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15506934

ABSTRACT

RecQ helicases are evolutionarily conserved enzymes required for the maintenance of genome stability. Mutations in three of the five known human RecQ helicase genes cause distinct clinical disorders that are characterized by genome instability and cancer predisposition. Recent studies have begun to reveal the cellular roles of RecQ helicases and how these enzymes may prevent tumorigenesis at the molecular level.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Genome , Neoplasms/prevention & control , Animals , Humans , Models, Genetic , RecQ Helicases , Saccharomyces cerevisiae/enzymology
10.
Nucleic Acids Res ; 30(10): 2124-30, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12000832

ABSTRACT

In mammalian cells, repair of the most abundant endogenous premutagenic lesion in DNA, 7,8-dihydro-8-oxoguanine (8-oxoG), is initiated by the bifunctional DNA glycosylase OGG1. By using purified human proteins, we have reconstituted repair of 8-oxoG lesions in DNA in vitro on a plasmid DNA substrate containing a single 8-oxoG residue. It is shown that efficient and complete repair requires only hOGG1, the AP endonuclease HAP1, DNA polymerase (Pol) beta and DNA ligase I. After glycosylase base removal, repair occurred through the AP lyase step of hOGG1 followed by removal of the 3'-terminal sugar phosphate by the 3'-diesterase activity of HAP1. Addition of PCNA had a slight stimulatory effect on repair. Fen1 or high concentrations of Pol beta were required to induce strand displacement DNA synthesis at incised 8-oxoG in the absence of DNA ligase. Fen1 induced Pol beta strand displacement DNA synthesis at HAP1-cleaved AP sites differently from that at gaps introduced by hOGG1/HAP1 at 8-oxoG sites. In the presence of DNA ligase I, the repair reaction at 8-oxoG was confined to 1 nt replacement, even in the presence of high levels of Pol beta and Fen1. Thus, the assembly of all the core proteins for 8-oxoG repair catalyses one major pathway that involves single nucleotide repair patches.


Subject(s)
DNA Repair , Guanine/metabolism , N-Glycosyl Hydrolases/metabolism , Base Sequence , Carbon-Oxygen Lyases/metabolism , DNA Ligase ATP , DNA Ligases/metabolism , DNA Polymerase beta/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , DNA-Binding Proteins/metabolism , DNA-Formamidopyrimidine Glycosylase , Endodeoxyribonucleases/metabolism , Flap Endonucleases , Guanine/analogs & derivatives , Humans , Oligonucleotides/genetics , Oligonucleotides/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Replication Protein C
11.
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
12.
Nucleic Acids Res ; 29(21): 4378-86, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11691925

ABSTRACT

Bloom's syndrome (BS) is a rare genetic disorder characterised by genomic instability and cancer susceptibility. BLM, the gene mutated in BS, encodes a member of the RecQ family of DNA helicases. Here, we identify hMLH1, which is involved in mismatch repair (MMR) and recombination, as a protein that directly interacts with BLM both in vivo and in vitro, and that the two proteins co-localise to discrete nuclear foci. The interaction between BLM and hMLH1 appears to have been evolutionarily conserved, as Sgs1p, the Saccharomyces cerevisiae homologue of BLM, interacts with yeast Mlh1p. However, cell extracts derived from BS patients show no obvious defects in MMR compared to wild-type- and BLM-complemented BS cell extracts. We conclude that the hMLH1-BLM interaction is not essential for post-replicative MMR, but, more likely, is required for some aspect of genetic recombination.


Subject(s)
Adenosine Triphosphatases/metabolism , Base Pair Mismatch , Bloom Syndrome , DNA Helicases/metabolism , DNA Repair , Neoplasm Proteins/metabolism , Protein Interaction Mapping , Adaptor Proteins, Signal Transducing , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Blotting, Far-Western , Carrier Proteins , Cell Line , Cell Nucleus/metabolism , Conserved Sequence , DNA Helicases/chemistry , DNA Helicases/genetics , Fungal Proteins/genetics , Fungal Proteins/metabolism , Humans , MutL Protein Homolog 1 , Mutation/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Nuclear Proteins/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , RecQ Helicases , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Two-Hybrid System Techniques
13.
EMBO J ; 20(22): 6530-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11707423

ABSTRACT

The major human AP endonuclease APE1 (HAP1, APEX, Ref1) initiates the repair of abasic sites generated either spontaneously, from attack of bases by free radicals, or during the course of the repair of damaged bases. APE1 therefore plays a central role in the base excision repair (BER) pathway. We report here that XRCC1, another essential protein involved in the maintenance of genome stability, physically interacts with APE1 and stimulates its enzymatic activities. A truncated form of APE1, lacking the first 35 amino acids, although catalytically proficient, loses the affinity for XRCC1 and is not stimulated by XRCC1. Chinese ovary cell lines mutated in XRCC1 have a diminished capacity to initiate the repair of AP sites. This defect is compensated by the expression of XRCC1. XRCC1, acting as both a scaffold and a modulator of the different activities involved in BER, would provide a physical link between the incision and sealing steps of the AP site repair process. The interaction described extends the coordinating role of XRCC1 to the initial step of the repair of DNA abasic sites.


Subject(s)
Carbon-Oxygen Lyases/metabolism , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Animals , Binding Sites , Blotting, Western , CHO Cells , Catalysis , Cricetinae , DNA/metabolism , DNA Primers/chemistry , DNA, Complementary/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase , Deoxyribonuclease IV (Phage T4-Induced) , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , HeLa Cells , Humans , Models, Biological , Mutagenesis, Site-Directed , Mutation , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Time Factors , Transcription, Genetic , Transfection , Two-Hybrid System Techniques , X-ray Repair Cross Complementing Protein 1
14.
Mol Cell Biol ; 21(21): 7150-62, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11585898

ABSTRACT

Deletion of the Saccharomyces cerevisiae TOP3 gene, encoding Top3p, leads to a slow-growth phenotype characterized by an accumulation of cells with a late S/G2 content of DNA (S. Gangloff, J. P. McDonald, C. Bendixen, L. Arthur, and R. Rothstein, Mol. Cell. Biol. 14:8391-8398, 1994). We have investigated the function of TOP3 during cell cycle progression and the molecular basis for the cell cycle delay seen in top3Delta strains. We show that top3Delta mutants exhibit a RAD24-dependent delay in the G2 phase, suggesting a possible role for Top3p in the resolution of abnormal DNA structures or DNA damage arising during S phase. Consistent with this notion, top3Delta strains are sensitive to killing by a variety of DNA-damaging agents, including UV light and the alkylating agent methyl methanesulfonate, and are partially defective in the intra-S-phase checkpoint that slows the rate of S-phase progression following exposure to DNA-damaging agents. This S-phase checkpoint defect is associated with a defect in phosphorylation of Rad53p, indicating that, in the absence of Top3p, the efficiency of sensing the existence of DNA damage or signaling to the Rad53 kinase is impaired. Consistent with a role for Top3p specifically during S phase, top3Delta mutants are sensitive to the replication inhibitor hydroxyurea, expression of the TOP3 mRNA is activated in late G1 phase, and DNA damage checkpoints operating outside of S phase are unaffected by deletion of TOP3. All of these phenotypic consequences of loss of Top3p function are at least partially suppressed by deletion of SGS1, the yeast homologue of the human Bloom's and Werner's syndrome genes. These data implicate Top3p and, by inference, Sgs1p in an S-phase-specific role in the cellular response to DNA damage. A model proposing a role for these proteins in S phase is presented.


Subject(s)
Cell Cycle Proteins , DNA Damage , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type I/physiology , Protein Serine-Threonine Kinases/metabolism , S Phase , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Alkylating Agents/pharmacology , Blotting, Northern , Blotting, Western , Cell Cycle , Checkpoint Kinase 2 , Dose-Response Relationship, Drug , Flow Cytometry , G2 Phase , Gene Deletion , Hydroxyurea/pharmacology , Methyl Methanesulfonate/pharmacology , Mitosis , Models, Biological , Mutation , Phenotype , Phosphorylation , Saccharomyces cerevisiae/enzymology , Time Factors , Ultraviolet Rays
15.
Cell Mol Life Sci ; 58(7): 894-901, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11497238

ABSTRACT

Maintenance of genomic stability relies on the efficient and accurate execution of DNA repair pathways, and is essential for cell viability and the prevention of cancer. Mutation of genes encoding RecQ helicases or topoisomerases gives rise to genomic instability through excessive recombination. Here, we review the recent biochemical and genetic evidence to indicate that these two classes of protein act in concert in a conserved pathway to maintain genomic stability by preventing inappropriate recombination.


Subject(s)
DNA Helicases/metabolism , DNA Replication , DNA Topoisomerases, Type I/metabolism , Recombination, Genetic , Animals , DNA Helicases/physiology , DNA Topoisomerases, Type I/physiology , Forecasting , Humans
16.
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
17.
Nucleic Acids Res ; 29(14): 2963-72, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11452021

ABSTRACT

In the budding yeast Saccharomyces cerevisiae the Srs2/RadH DNA helicase promotes survival after ultraviolet (UV) irradiation, and has been implicated in DNA repair, recombination and checkpoint signalling following DNA damage. A second helicase, Sgs1, is the S.cerevisiae homologue of the human BLM and WRN proteins, which are defective in cancer predisposition and/or premature ageing syndromes. Saccharomyces cerevisiae cells lacking both Srs2 and Sgs1 exhibit a severe growth defect. We have identified an Srs2 orthologue in the fission yeast Schizosaccharomyces pombe, and have investigated its role in responses to UV irradiation and inhibition of DNA replication. Deletion of fission yeast srs2 caused spontaneous hyper-recombination and UV sensitivity, and simultaneous deletion of the SGS1 homologue rqh1 caused a severe growth defect reminiscent of that seen in the equivalent S.cerevisiae mutant. However, unlike in budding yeast, inactivation of the homologous recombination pathway did not suppress this growth defect. Indeed, the homologous recombination pathway was required for maintenance of normal fission yeast viability in the absence of Srs2, and loss of homologous recombination and loss of Srs2 contributed additively to UV sensitivity. We conclude that Srs2 plays related, but not identical, roles in the two yeast species.


Subject(s)
DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins , Saccharomyces cerevisiae Proteins , Schizosaccharomyces pombe Proteins , Schizosaccharomyces/genetics , Amino Acid Sequence , Cell Division/drug effects , Cell Division/genetics , Cell Division/radiation effects , DNA Helicases/genetics , DNA Repair , DNA Topoisomerases, Type I/genetics , Fungal Proteins/genetics , Gene Deletion , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Genes, Lethal , Hydroxyurea/pharmacology , Molecular Sequence Data , Phenotype , Rad51 Recombinase , Recombination, Genetic , Schizosaccharomyces/enzymology , Sequence Homology, Amino Acid , Ultraviolet Rays
18.
Br J Cancer ; 85(2): 261-5, 2001 Jul 20.
Article in English | MEDLINE | ID: mdl-11461087

ABSTRACT

Bloom's syndrome (BS) is an autosomal recessive disorder associated with a predisposition to cancers of all types. Cells from BS sufferers display extreme genomic instability. The BS gene product, BLM, is a 159 kDa DNA helicase enzyme belonging to the RecQ family. Here, we have analysed the distribution of BLM in normal and tumour tissues from humans using a recently characterized, specific monoclonal antibody. BLM was found to be localized to nuclei in normal lymphoid tissues, but was largely absent from other normal tissues analysed with the exception of the proliferating compartment of certain tissues. In contrast, expression of BLM was observed in a variety of tumours of both lymphoid and epithelial origin. A strong correlation was observed between expression of BLM and the proliferative status of cells, as determined by staining for markers of cell proliferation (PCNA and Ki67). We conclude that BLM is a proliferation marker in normal and neoplastic cells in vivo, and, as a consequence, is expressed at a higher level in tumours than in normal quiescent tissues.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , Neoplasms/metabolism , Adenosine Triphosphatases/immunology , Antibodies, Monoclonal/immunology , DNA Helicases/immunology , Humans , Neoplasms/classification , RecQ Helicases
19.
Nucleic Acids Res ; 29(15): 3172-80, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11470874

ABSTRACT

In higher eukaryotes, the integration of signals triggered in response to certain types of stress can result in programmed cell death. Central to these events is the sequential activation of a cascade of proteinases known as caspases. The final activated effector caspases of this cascade digest a number of cellular proteins, in some cases increasing their enzymatic activity, in others destroying their function. Of the proteins shown to be targets for caspase-mediated proteolysis, a surprisingly large proportion are proteins involved in the signalling or repair of DNA damage. Here we investigate whether BLM, the product of the gene mutated in Bloom's syndrome, a human autosomal disease characterised by cancer predisposition and sunlight sensitivity, is cleaved during apoptosis. BLM interacts with topoisomerase IIIalpha and has been proposed to play an important role in maintaining genomic integrity through its roles in DNA repair and replication. We show that BLM is cleaved during apoptosis by caspase-3 and reveal that the main cleavage site is located at the junction between the N-terminal and central helicase domains of BLM. Proteolytic cleavage by caspase-3 produces a 120 kDa fragment, which contains the intact helicase domain and three smaller fragments, the relative amounts of which depend on time of incubation with caspase-3. The 120 kDa fragment retains the helicase activity of the intact BLM protein. However, its interaction with topoisomerase IIIalpha is severely impaired. Since the BLM-topoisomerase interaction is believed to be necessary for many of the replication and recombination functions of BLM, we suggest that caspase-3 cleavage of BLM could alter the localisation and/or function of BLM and that these changes may be important in the process of apoptosis.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis , Caspases/metabolism , DNA Helicases/metabolism , DNA Topoisomerases, Type I/metabolism , Adenosine Triphosphatases/chemistry , Apoptosis/drug effects , Bloom Syndrome/enzymology , Caspase 3 , Cycloheximide/pharmacology , DNA Helicases/chemistry , Etoposide/pharmacology , HL-60 Cells , HeLa Cells , Humans , Models, Biological , Molecular Weight , Protein Binding , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary , RecQ Helicases , Tumor Necrosis Factor-alpha/pharmacology
20.
Biochem Soc Trans ; 29(Pt 2): 201-4, 2001 May.
Article in English | MEDLINE | ID: mdl-11356154

ABSTRACT

The RecQ family of DNA helicases has members in all organisms analysed. In humans, defects in three family members are associated with disease conditions: BLM is defective in Bloom's syndrome, WRN in Werner's syndrome and RTS in Rothmund-Thomson syndrome. In each case, cells from affected individuals show inherent genomic instability. The focus of our work is the Bloom's syndrome gene and its product, BLM. Here, we review the latest information concerning the roles of BLM in the maintenance of genome integrity.


Subject(s)
Adenosine Triphosphatases/metabolism , Bloom Syndrome/enzymology , DNA Helicases/metabolism , Genome, Human , Adenosine Triphosphatases/chemistry , Chromosome Aberrations/genetics , DNA Helicases/chemistry , Humans , Phenotype , Protein Binding , Protein Structure, Tertiary , Protein Transport , RecQ Helicases , Recombination, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...