Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Br J Cancer ; 103(12): 1875-84, 2010 Dec 07.
Article in English | MEDLINE | ID: mdl-21063410

ABSTRACT

BACKGROUND: defective DNA repair has a causal role in hereditary colorectal cancer (CRC). Defects in the base excision repair gene MUTYH are responsible for MUTYH-associated polyposis and CRC predisposition as an autosomal recessive trait. Numerous reports have suggested MUTYH mono-allelic variants to be low penetrance risk alleles. We report a large collaborative meta-analysis to assess and refine CRC risk estimates associated with bi-allelic and mono-allelic MUTYH variants and investigate age and sex influence on risk. METHODS: MUTYH genotype data were included from 20 565 cases and 15 524 controls. Three logistic regression models were tested: a crude model; adjusted for age and sex; adjusted for age, sex and study. RESULTS: all three models produced very similar results. MUTYH bi-allelic carriers demonstrated a 28-fold increase in risk (95% confidence interval (CI): 6.95-115). Significant bi-allelic effects were also observed for G396D and Y179C/G396D compound heterozygotes and a marginal mono-allelic effect for variant Y179C (odds ratio (OR)=1.34; 95% CI: 1.00-1.80). A pooled meta-analysis of all published and unpublished datasets submitted showed bi-allelic effects for MUTYH, G396D and Y179C (OR=10.8, 95% CI: 5.02-23.2; OR=6.47, 95% CI: 2.33-18.0; OR=3.35, 95% CI: 1.14-9.89) and marginal mono-allelic effect for variants MUTYH (OR=1.16, 95% CI: 1.00-1.34) and Y179C alone (OR=1.34, 95% CI: 1.01-1.77). CONCLUSIONS: overall, this large study refines estimates of disease risk associated with mono-allelic and bi-allelic MUTYH carriers.


Subject(s)
Colorectal Neoplasms/genetics , DNA Glycosylases/genetics , Adult , Aged , Colorectal Neoplasms/etiology , Female , Genetic Predisposition to Disease , Humans , Male , Middle Aged , Mutation , Risk Factors
2.
Mech Ageing Dev ; 129(11): 681-91, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19238688

ABSTRACT

Human cells express five DNA helicases that are paralogs of Escherichia coli RecQ and which constitute the family of human RecQ helicases. Disease-causing mutations in three of these five human DNA helicases, BLM, WRN, and RECQL4, cause rare severe human genetic diseases with distinct clinical phenotypes characterized by developmental defects, skin abnormalities, genomic instability, and cancer susceptibility. Although biochemical and genetic evidence support roles for all five human RecQ helicases in DNA replication, DNA recombination, and the biological responses to DNA damage, many questions concerning the various functions of the human RecQ helicases remain unanswered. Researchers investigating human and non-human RecQ helicases held a workshop on May 27-28, 2008, at the University of Chicago Gleacher Center, during which they shared insights, discussed recent progress in understanding the biochemistry, biology, and genetics of the RecQ helicases, and developed research strategies that might lead to therapeutic approaches to the human diseases that result from mutations in RecQ helicase genes. Some workshop sessions were held jointly with members of a recently formed advocacy and support group for persons with Bloom's syndrome and their families. This report describes the outcomes and main discussion points of the workshop.


Subject(s)
Bloom Syndrome/enzymology , RecQ Helicases/metabolism , Aging/genetics , Aging/metabolism , Animals , Biomedical Research , Bloom Syndrome/genetics , DNA Damage , DNA Replication , Humans , Mutation , Neoplasms/enzymology , Neoplasms/genetics , RecQ Helicases/deficiency , RecQ Helicases/genetics , Recombination, Genetic , Self-Help Groups
3.
Am J Hum Genet ; 71(6): 1395-412, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12454801

ABSTRACT

Hereditary nonpolyposis colorectal cancer (HNPCC) is caused by mutations in the mismatch-repair genes. We report here the identification and characterization of a founder mutation in MSH2 in the Ashkenazi Jewish population. We identified a nucleotide substitution, MSH2*1906G-->C, which results in a substitution of proline for alanine at codon 636 in the MSH2 protein. This allele was identified in 15 unrelated Ashkenazi Jewish families with HNPCC, most of which meet the Amsterdam criteria. Genotype analysis of 18 polymorphic loci within and flanking MSH2 suggested a single origin for the mutation. All colorectal cancers tested showed microsatellite instability and absence of MSH2 protein, by immunohistochemical analysis. In an analysis of a population-based incident series of 686 Ashkenazi Jews from Israel who have colorectal cancer, we identified 3 (0.44%) mutation carriers. Persons with a family history of colorectal or endometrial cancer were more likely to carry the mutation than were those without such a family history (P=.042), and those with colorectal cancer who carried the mutation were, on average, younger than affected individuals who did not carry it (P=.033). The mutation was not detected in either 566 unaffected Ashkenazi Jews from Israel or 1,022 control individuals from New York. In hospital-based series, the 1906C allele was identified in 5/463 Ashkenazi Jews with colorectal cancer, in 2/197 with endometrial cancer, and in 0/83 with ovarian cancer. When families identified by family history and in case series are included, 25 apparently unrelated Ashkenazi Jewish families have been found to harbor this mutation. Although this pathogenic mutation is not frequent in the Ashkenazi Jewish population (accounting for 2%-3% of colorectal cancer in those whose age at diagnosis is <60 years), it is highly penetrant and accounts for approximately one-third of HNPCC in Ashkenazi Jewish families that fulfill the Amsterdam criteria.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA-Binding Proteins , Founder Effect , Genetic Predisposition to Disease , Jews/genetics , Point Mutation/genetics , Proto-Oncogene Proteins/genetics , Alanine/genetics , Case-Control Studies , Chromosomes, Human, Pair 2/genetics , Crystallography, X-Ray , Female , Gene Frequency/genetics , Haplotypes/genetics , Heterozygote , Humans , Israel , Male , Microsatellite Repeats/genetics , MutS Homolog 2 Protein , Mutation, Missense/genetics , Neoplasms/genetics , Pedigree , Polymorphism, Genetic/genetics , Proline/genetics , Protein Conformation , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/chemistry
5.
Hum Mol Genet ; 10(12): 1287-98, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11406610

ABSTRACT

The genomic instability of persons with Bloom's syndrome (BS) features particularly an increased number of sister-chromatid exchanges (SCEs). The primary cause of the genomic instability is mutation at BLM, which encodes a DNA helicase of the RecQ family. BLM interacts with Topoisomerase IIIalpha (Topo IIIalpha), and both BLM and Topo IIIalpha localize to the nuclear organelles referred to as the promyelocytic leukemia protein (PML) nuclear bodies. In this study we show, by analysis of cells that express various deletion constructs of green fluorescent protein (GFP)-tagged BLM, that the first 133 amino acids of BLM are necessary and sufficient for interaction between Topo IIIalpha and BLM. The Topo IIIalpha-interaction domain of BLM is not required for BLM's localization to the PML nuclear bodies; in contrast, Topo IIIalpha is recruited to the PML nuclear bodies via its interaction with BLM. Expression of a full-length BLM (amino acids 1-1417) in BS cells can correct their high SCEs to normal levels, whereas expression of a BLM fragment that lacks the Topo IIIalpha interaction domain (amino acids 133-1417) results in intermediate SCE levels. The deficiency of amino acids 133-1417 in the reduction of SCEs was not explained by a defect in DNA helicase activity, because immunoprecipitated 133-1417 protein had 4-fold higher activity than GFP-BLM. The data implicate the BLM-Topo IIIalpha complex in the regulation of recombination in somatic cells.


Subject(s)
Adenosine Triphosphatases/metabolism , Bloom Syndrome/enzymology , Bloom Syndrome/genetics , DNA Helicases/metabolism , DNA Topoisomerases, Type I/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Binding Sites , Bloom Syndrome/metabolism , Cell Line, Transformed , DNA Helicases/chemistry , DNA Helicases/genetics , DNA Topoisomerases, Type I/genetics , Gene Expression Regulation , HeLa Cells , Humans , Phenotype , Protein Structure, Tertiary , RecQ Helicases , Sister Chromatid Exchange , Tumor Cells, Cultured
6.
J Biol Chem ; 276(35): 32948-55, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11399766

ABSTRACT

The Bloom syndrome (BS) protein, BLM, is a member of the RecQ DNA helicase family that also includes the Werner syndrome protein, WRN. Inherited mutations in these proteins are associated with cancer predisposition of these patients. We recently discovered that cells from Werner syndrome patients displayed a deficiency in p53-mediated apoptosis and WRN binds to p53. Here, we report that analogous to WRN, BLM also binds to p53 in vivo and in vitro, and the C-terminal domain of p53 is responsible for the interaction. p53-mediated apoptosis is defective in BS fibroblasts and can be rescued by expression of the normal BLM gene. Moreover, lymphoblastoid cell lines (LCLs) derived from BS donors are resistant to both gamma-radiation and doxorubicin-induced cell killing, and sensitivity can be restored by the stable expression of normal BLM. In contrast, BS cells have a normal Fas-mediated apoptosis, and in response to DNA damage normal accumulation of p53, normal induction of p53 responsive genes, and normal G(1)-S and G(2)-M cell cycle arrest. BLM localizes to nuclear foci referred to as PML nuclear bodies (NBs). Cells from Li-Fraumeni syndrome patients carrying p53 germline mutations and LCLs lacking a functional p53 have a decreased accumulation of BLM in NBs, whereas isogenic lines with functional p53 exhibit normal accumulation. Certain BLM mutants (C1055S or Delta133-237) that have a reduced ability to localize to the NBs when expressed in normal cells can impair the localization of wild type BLM to NBs and block p53-mediated apoptosis, suggesting a dominant-negative effect. Taken together, our results indicate both a novel mechanism of p53 function by which p53 mediates nuclear trafficking of BLM to NBs and the cooperation of p53 and BLM to induce apoptosis.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Apoptosis/physiology , Bloom Syndrome/enzymology , Cell Cycle/physiology , DNA Damage , DNA Helicases/chemistry , DNA Helicases/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Apoptosis/radiation effects , Binding Sites , Bloom Syndrome/genetics , Cell Line , Cell Nucleus/physiology , Cell Survival , Dose-Response Relationship, Radiation , Fibroblasts/cytology , Fibroblasts/physiology , Fibroblasts/radiation effects , Fluorescent Antibody Technique, Indirect , Gamma Rays , Genes, Reporter , Humans , RecQ Helicases , Recombinant Proteins/metabolism , Reference Values , Transfection
7.
J Cell Biol ; 153(2): 367-80, 2001 Apr 16.
Article in English | MEDLINE | ID: mdl-11309417

ABSTRACT

Bloom syndrome (BS) is an autosomal recessive disorder characterized by a high incidence of cancer and genomic instability. BLM, the protein defective in BS, is a RecQ-like helicase, presumed to function in DNA replication, recombination, or repair. BLM localizes to promyelocytic leukemia protein (PML) nuclear bodies and is expressed during late S and G2. We show, in normal human cells, that the recombination/repair proteins hRAD51 and replication protein (RP)-A assembled with BLM into a fraction of PML bodies during late S/G2. Biochemical experiments suggested that BLM resides in a nuclear matrix-bound complex in which association with hRAD51 may be direct. DNA-damaging agents that cause double strand breaks and a G2 delay induced BLM by a p53- and ataxia-telangiectasia mutated independent mechanism. This induction depended on the G2 delay, because it failed to occur when G2 was prevented or bypassed. It coincided with the appearance of foci containing BLM, PML, hRAD51 and RP-A, which resembled ionizing radiation-induced foci. After radiation, foci containing BLM and PML formed at sites of single-stranded DNA and presumptive repair in normal cells, but not in cells with defective PML. Our findings suggest that BLM is part of a dynamic nuclear matrix-based complex that requires PML and functions during G2 in undamaged cells and recombinational repair after DNA damage.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle/physiology , Cell Nucleus/metabolism , DNA Damage , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Adenosine Triphosphatases/genetics , Bloom Syndrome/genetics , Blotting, Western , Cell Fractionation , Cells, Cultured , DNA Helicases/genetics , DNA Repair/physiology , DNA-Binding Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/radiation effects , Flow Cytometry , Humans , Microscopy, Fluorescence , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Proteins/metabolism , Rad51 Recombinase , RecQ Helicases , Replication Protein A , Transcription Factors/metabolism , Tubulin/metabolism , Tumor Suppressor Proteins , X-Rays
8.
Hum Genet ; 108(2): 167-73, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11281456

ABSTRACT

A unique and constant feature of Bloom syndrome (BS) cells is an excessive rate of sister-chromatid exchange (SCE). However, in approximately 20% of persons with typical BS, mosaicism is observed in which a proportion of lymphocytes (usually a small one) exhibits a low-SCE rate. Persons with such mosaicism predominantly are genetic compounds for mutation at BLM, and the low-SCE lymphocytes are the progeny of a precursor cell in which intragenic recombination between the two sites of BLM mutation had generated a normal allele. Very exceptionally, however, persons with BS who exhibit mosaicism are homozygous for the causative mutation. In two such exceptional homozygous persons studied here, back mutation has been demonstrated: one person constitutionally was homozygous for the mutation 1544insA and the other for the mutation 2702G-->A. Revertant (low-SCE) lymphoblastoid cells in each person were heterozygous for their mutations, i.e., a normal allele was now present. The normal alleles must have arisen by back mutation in a precursor cell, in one person by the deletion of an A base and, in the other, the nucleotide substitution of a G base for an A base. Thus, back mutation now becomes, together with intragenic recombination, an important genetic mechanism to consider when explaining examples of a reversion of somatic cells to "normal" in persons with a genetically determined abnormal phenotype.


Subject(s)
Bloom Syndrome/genetics , Mutation , Bloom Syndrome/pathology , Heterozygote , Homozygote , Humans , Phenotype , Sister Chromatid Exchange
9.
Biochemistry ; 39(47): 14617-25, 2000 Nov 28.
Article in English | MEDLINE | ID: mdl-11087418

ABSTRACT

Bloom syndrome is a rare autosomal disorder characterized by predisposition to cancer and genomic instability. BLM, the structural gene mutated in individuals with the disorder, encodes a DNA helicase belonging to the RecQ family of helicases. These helicases have been established to serve roles in both promoting and preventing recombination. Mounting evidence has implicated a function for BLM during DNA replication; specifically, BLM might be involved in rescuing stalled or collapsed replication forks by a recombination-based mechanism. We have tested this idea by examining the binding and melting activity of BLM on oligonucleotide substrates containing D-loops, DNA structures that model the presumed initial intermediate formed during homologous recombination. We find that BLM preferentially melts those D-loops that are formed more favorably by the strand exchange protein Rad51, but whose polarity could be less favorable for enabling restoration of an active replication fork. We propose a model in which BLM selectively dissociates recombination intermediates likely to be unfavorable for recombination-promoted replication.


Subject(s)
Adenosine Triphosphatases/metabolism , Bloom Syndrome/enzymology , Bloom Syndrome/genetics , DNA Damage , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Nucleic Acid Conformation , Adenosine Triphosphatases/chemistry , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Humans , Models, Genetic , Nucleic Acid Heteroduplexes/metabolism , Protein Binding , Rad51 Recombinase , RecQ Helicases , Recombination, Genetic , Substrate Specificity
10.
Oncogene ; 19(41): 4764-72, 2000 Sep 28.
Article in English | MEDLINE | ID: mdl-11032027

ABSTRACT

Three human RecQ DNA helicases, WRN, BLM and RTS, are involved in the genetic disorders associated with genomic instability and a high incidence of cancer. RecQL1 and RecQL5 also belong to the human RecQ helicase family, but their correlation with genetic disorders, if any, is unknown. We report here that in human B cells transformed by Epstein-Barr virus (EBV), human fibroblasts and umbilical endothelial cells transformed by simian virus 40, the expression of WRN, BLM, RTS and RecQL1 was sharply up-regulated. In B cells this expression was stimulated within 5-40 h by the tumor promoting agent phorbol myristic acetate (PMA). Interestingly, RecQL5beta, an alternative splicing product of RecQL5 with a nuclear localization signal, is expressed in resting B cells without significant modulation of its synthesis by EBV or PMA, suggesting it has a role in resting cells. We also roughly determined the number of copies per cell for the five RecQ helicase in B cells. In addition, levels of the different RecQ helicases are modulated in different ways during the cell cycle of actively proliferating fibroblasts and umbilical endothelial cells. Our results support the view that the levels of WRN, BLM, RTS and RecQL1 are differentially up-regulated to guarantee genomic stability in cells that are transformed or actively proliferating.


Subject(s)
Adenosine Triphosphatases/biosynthesis , Cell Cycle/physiology , Cell Transformation, Viral/genetics , DNA Helicases/biosynthesis , Gene Expression Regulation, Enzymologic , Werner Syndrome/enzymology , Adenosine Triphosphatases/genetics , Alternative Splicing , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , B-Lymphocytes/virology , Cell Division , Cell Line, Transformed/enzymology , DNA Helicases/deficiency , DNA Helicases/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Enzyme Induction , Exodeoxyribonucleases , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Herpesvirus 4, Human/physiology , Humans , RecQ Helicases , Simian virus 40/physiology , Tetradecanoylphorbol Acetate/pharmacology , Umbilical Veins/cytology , Werner Syndrome/blood , Werner Syndrome/genetics , Werner Syndrome Helicase
11.
Nucleic Acids Res ; 28(12): 2396-405, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10871373

ABSTRACT

Werner's syndrome (WS) is an autosomal recessive disorder in humans characterized by the premature development of a partial array of age-associated pathologies. WRN, the gene defective in WS, encodes a 1432 amino acid protein (hWRN) with intrinsic 3'-->5' DNA helicase activity. We recently showed that hWRN is also a 3'-->5' exonuclease. Here, we further characterize the hWRN exonuclease. hWRN efficiently degraded the 3' recessed strands of double-stranded DNA or a DNA-RNA heteroduplex. It had little or no activity on blunt-ended DNA, DNA with a 3' protruding strand, or single-stranded DNA. The hWRN exonuclease efficiently removed a mismatched nucleotide at a 3' recessed terminus, and was capable of initiating DNA degradation from a 12-nt gap, or a nick. We further show that the mouse WRN (mWRN) is also a 3'-->5' exonuclease, with substrate specificity similar to that of hWRN. Finally, we show that hWRN forms a trimer and interacts with the proliferating cell nuclear antigen in vitro. These findings provide new data on the biochemical activities of WRN that may help elucidate its role(s) in DNA metabolism.


Subject(s)
DNA Helicases/metabolism , Exodeoxyribonucleases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Helicases/chemistry , DNA Helicases/genetics , Exodeoxyribonuclease V , Humans , Mice , Molecular Sequence Data , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/metabolism , RecQ Helicases , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Werner Syndrome/enzymology , Werner Syndrome/genetics , Werner Syndrome Helicase
12.
Cancer Res ; 60(5): 1162-7, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10728666

ABSTRACT

Bloom syndrome (BS) is characterized by genomic instability and cancer susceptibility caused by defects in BLM, a DNA helicase of the RecQ-family (J. German and N. A. Ellis, The Genetic Basis of Human Cancer, pp. 301-316, 1998). RecQ helicases and topoisomerase III proteins interact physically and functionally in yeast (S. Gangloff et al., Mol. Cell. Biol., 14: 8391-8398, 1994) and in Escherichia coli can function together to enable passage of double-stranded DNA (F. G. Harmon et al., Mol. Cell, 3: 611-620, 1999). We demonstrate in somatic and meiotic human cells an association between BLM and topoisomerase IIIalpha. These proteins colocalize in promyelocytic leukemia protein nuclear bodies, and this localization is disrupted in BS cells. Thus, mechanisms by which RecQ helicases and topoisomerase III proteins cooperate to maintain genomic stability in model organisms likely apply to humans.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , DNA Topoisomerases, Type I/metabolism , Meiosis , Adenosine Triphosphatases/genetics , DNA Helicases/genetics , DNA Topoisomerases, Type I/genetics , Gene Expression Regulation , Humans , RecQ Helicases , Tumor Cells, Cultured
13.
Cytogenet Cell Genet ; 91(1-4): 217-23, 2000.
Article in English | MEDLINE | ID: mdl-11173860

ABSTRACT

BLM, the protein encoded by the gene mutated in Bloom's syndrome (BS), is a phylogenetically highly conserved DNA helicase that varies in amount and distribution in the nucleus during the cell-division cycle. It is undetectable in many cells as they emerge from mitosis but becomes abundant during G(1) and remains so throughout S, G(2), and mitosis. BLM is widely distributed throughout the nucleus but at certain times also becomes concentrated in foci that vary in number and size. It co-localizes transitorily with replication protein A (RPA) and promyelocytic leukemia protein (PML) nuclear bodies, and at times it enters the nucleolus. The observations support the hypothesis that BLM is distributed variously about the nucleus to manipulate DNA in some, very possibly several, nucleic acid transactions, when and where they take place. The specific transaction(s) remain to be identified. Although absence from the nucleus of functional BLM - the situation in BS - obviously is not lethal in the human, other helicases would appear to be unable to substitute for it completely, witness the hypermutability and hyperrecombinability of BS cells.


Subject(s)
Adenosine Triphosphatases/metabolism , Bloom Syndrome/genetics , Cell Cycle , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Adenosine Triphosphatases/genetics , B-Lymphocytes , Bloom Syndrome/metabolism , Cell Line, Transformed , Cell Nucleolus/chemistry , Cell Nucleolus/metabolism , Cells, Cultured , DNA Helicases/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Fibroblasts , Fluorescent Antibody Technique , HL-60 Cells , Humans , Male , Nuclear Proteins/genetics , Protein Subunits , Protein Transport , RecQ Helicases , Replication Protein A
14.
Article in English | MEDLINE | ID: mdl-11701636

ABSTRACT

DNA helicases are a highly conserved group of enzymes that unwind DNA. They function in all processes in which access to single-stranded DNA is required, including DNA replication, DNA repair and recombination, and transcription of RNA. Defects in helicases functioning in one or more of these processes can result in characteristic human genetic disorders in which genomic instability and predisposition to cancer are common features. So far, different helicase genes have been found mutated in six such disorders. Mutations in XPB and XPD can result in xeroderma pigmentosum, Cockayne syndrome, or trichothiodystrophy. Mutations in the RecQ-like genes BLM, WRN, and RECQL4 can result in Bloom syndrome, Werner syndrome, and Rothmund-Thomson syndrome, respectively. Because XPB and XPD function in both nucleotide excision repair and transcription initiation, the cellular phenotypes associated with a deficiency of each one of them include failure to repair mutagenic DNA lesions and defects in the recovery of RNA transcription after UV irradiation. The functions of the RecQ-like genes are unknown; however, a growing body of evidence points to a function in restarting DNA replication after the replication fork has become stalled. The genomic instability associated with mutations in the RecQ-like genes includes spontaneous chromosome instability and elevated mutation rates. Mouse models for nearly all of these entities have been developed, and these should help explain the widely different clinical features that are associated with helicase mutations.


Subject(s)
DNA Helicases/genetics , Genetic Diseases, Inborn/enzymology , Genetic Diseases, Inborn/genetics , Saccharomyces cerevisiae Proteins , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Transcription Factors, TFII , Amino Acid Sequence , Animals , Bloom Syndrome/genetics , DNA Helicases/chemistry , DNA Helicases/metabolism , DNA Repair , DNA Replication , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Phenotype , Recombination, Genetic , Rothmund-Thomson Syndrome/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Transcription Factor TFIIH , Transcription Factors/genetics , Transcription Factors/metabolism , Werner Syndrome/genetics
15.
Protein Expr Purif ; 17(2): 239-48, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545272

ABSTRACT

BLM is a DNA helicase encoded by a gene which is mutated in persons with Bloom's syndrome. The protein is a member of the RecQ subfamily of helicases and contains a central domain constituted by the seven motifs conserved in all helicases. In contrast, the N-terminal portion of BLM lacks similarity to any other known proteins or motifs. We have expressed the first 431 amino acids of this domain as a fusion to a hexahistidine tag (BLM N431) in Escherichia coli. A method of purification was developed which involves elution from Ni-NTA resin in imidazole and EDTA, followed by treatment with DTT and gel filtration on Sephacryl-300. The treatment with EDTA and DTT prevents and disrupts aggregation of BLM N431. The purified protein appears to form hexamers and dodecamers, suggesting that the N-terminal domain of BLM is involved in the organization of the quaternary structure of BLM.


Subject(s)
Adenosine Triphosphatases/isolation & purification , DNA Helicases/isolation & purification , Histidine/metabolism , Oligopeptides/isolation & purification , Adenosine Triphosphatases/genetics , Amino Acid Motifs , Animals , Bloom Syndrome , Chelating Agents , Chromatography, Affinity/methods , Chromatography, Gel/methods , DNA Helicases/genetics , Edetic Acid/pharmacology , Escherichia coli/genetics , Gene Expression , Humans , Molecular Probes , Nickel , Nitrilotriacetic Acid/analogs & derivatives , Nitrilotriacetic Acid/pharmacology , Oligopeptides/drug effects , Organometallic Compounds/pharmacology , RecQ Helicases , Solubility
16.
Am J Hum Genet ; 65(5): 1368-74, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10521302

ABSTRACT

The gene BLM, mutated in Bloom syndrome (BS), encodes the nuclear protein BLM, which when absent, as it is from most BS cells, results in genomic instability. A manifestation of this instability is an excessive rate of sister-chromatid exchange (SCE). Here we describe the effects on this abnormal cellular phenotype of stable transfection of normal BLM cDNAs into two types of BS cells, SV40-transformed fibroblasts and Epstein-Barr virus (EBV)-transformed lymphoblastoid cells. Clones of BLM-transfected fibroblasts produced normal amounts of BLM by western blot analysis and displayed a normal nuclear localization of the protein by immunofluorescence microscopy. They had a mean of 24 SCEs/46 chromosomes, in contrast to the mean of 69 SCEs in controls transfected only with the vector. BLM-transfected fibroblast clones that expressed highest levels of the BLM protein had lowest levels of SCE. The lymphoblastoid cells transfected with BLM had SCE frequencies of 22 and 42 in two separate experiments in which two different selectable markers were used, in contrast to 57 and 58 in vector-transfected cells; in this type cell, however, the BLM protein was below the level detectable by western blot analysis. These experiments prove that BLM cDNA encodes a functional protein capable of restoring to or toward normal the uniquely characteristic high-SCE phenotype of BS cells.


Subject(s)
Adenosine Triphosphatases/genetics , Bloom Syndrome/genetics , DNA Helicases/genetics , Sister Chromatid Exchange/genetics , Adenosine Triphosphatases/metabolism , Blotting, Western , Cells, Cultured , DNA Helicases/metabolism , Fibroblasts , Fluorescent Antibody Technique , Herpesvirus 4, Human/genetics , Humans , Lymphocytes , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , RecQ Helicases , Simian virus 40/genetics , Transfection , Transformation, Genetic
17.
Mol Biol Cell ; 10(3): 665-76, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10069810

ABSTRACT

Bloom syndrome (BS) is a rare autosomal recessive disorder characterized by growth deficiency, immunodeficiency, genomic instability, and the early development of cancers of many types. BLM, the protein encoded by BLM, the gene mutated in BS, is localized in nuclear foci and absent from BS cells. BLM encodes a DNA helicase, and proteins from three missense alleles lack displacement activity. BLM transfected into BS cells reduces the frequency of sister chromatid exchanges and restores BLM in the nucleus. Missense alleles fail to reduce the sister chromatid exchanges in transfected BS cells or restore the normal nuclear pattern. BLM complements a phenotype of a Saccharomyces cerevisiae sgs1 top3 strain, and the missense alleles do not. This work demonstrates the importance of the enzymatic activity of BLM for its function and nuclear localization pattern.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Bloom Syndrome/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Adenosine Triphosphatases/isolation & purification , Bloom Syndrome/metabolism , Child, Preschool , DNA Helicases/isolation & purification , DNA, Complementary/metabolism , Female , Gene Expression Regulation , Genetic Complementation Test , Humans , Male , Mutation , Phenotype , RecQ Helicases , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins
18.
Oncogene ; 18(56): 7941-7, 1999 Dec 23.
Article in English | MEDLINE | ID: mdl-10637504

ABSTRACT

The PML gene of acute promyelocytic leukemia (APL) encodes a cell-growth and tumor suppressor. PML localizes to discrete nuclear bodies (NBs) that are disrupted in APL cells. The Bloom syndrome gene BLM encodes a RecQ DNA helicase, whose absence from the cell results in genomic instability epitomized by high levels of sister-chromatid exchange (SCE) and cancer predisposition. We show here that BLM co-localizes with PML to the NB. In cells from persons with Bloom syndrome the localization of PML is unperturbed, whereas in APL cells carrying the PML-RARalpha oncoprotein, both PML and BLM are delocalized from the NB into microspeckled nuclear regions. Treatment with retinoic acid (RA) induces the relocalization of both proteins to the NB. In primary PML-/- cells, BLM fails to accumulate in the NB. Strikingly, in PML-/- cells the frequency of SCEs is increased relative to PML+/+ cells. These data demonstrate that BLM is a constituent of the NB and that PML is required for its accumulation in these nuclear domains and for the normal function of BLM. Thus, our findings suggest a role for BLM in APL pathogenesis and implicate the PML NB in the maintenance of genomic stability.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Nucleus/metabolism , DNA Helicases/genetics , Leukemia, Promyelocytic, Acute/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Adenosine Triphosphatases/metabolism , Animals , Bloom Syndrome/genetics , Cell Line , Cell Nucleus/drug effects , Codon, Terminator , DNA Helicases/metabolism , Fluorescent Antibody Technique, Indirect , HL-60 Cells , HeLa Cells , Homozygote , Humans , Keratinocytes/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , RecQ Helicases , Sister Chromatid Exchange , Transfection , Tretinoin/pharmacology , Tumor Suppressor Proteins , U937 Cells
19.
Am J Hum Genet ; 63(6): 1685-93, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9837821

ABSTRACT

Bloom syndrome (BS) is more frequent in the Ashkenazic Jewish population than in any other. There the predominant mutation, referred to as "blmAsh," is a 6-bp deletion and 7-bp insertion at nucleotide position 2281 in the BLM cDNA. Using a convenient PCR assay, we have identified blmAsh on 58 of 60 chromosomes transmitted by Ashkenazic parents to persons with BS. In contrast, in 91 unrelated non-Ashkenazic persons with BS whom we examined, blmAsh was identified only in 5, these coming from Spanish-speaking Christian families from the southwestern United States, Mexico, or El Salvador. These data, along with haplotype analyses, show that blmAsh was independently established through a founder effect in Ashkenazic Jews and in immigrants to formerly Spanish colonies. This striking observation underscores the complexity of Jewish history and demonstrates the importance of migration and genetic drift in the formation of human populations.


Subject(s)
Adenosine Triphosphatases/genetics , Bloom Syndrome/genetics , DNA Helicases/genetics , Jews/genetics , Mutation , Alleles , Bloom Syndrome/ethnology , Christianity , Chromosomes, Human , Consanguinity , El Salvador , Founder Effect , Gene Frequency , Genotype , Haplotypes , Humans , Mexico , Polymerase Chain Reaction , Polymorphism, Genetic , RecQ Helicases , Spain/ethnology , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...