Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Publication year range
1.
Nat Genet ; 17(1): 100-3, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9288107

ABSTRACT

Werner syndrome (WS) is an uncommon autosomal recessive disorder characterized by premature aging. The clinical manifestations of WS, including atherosclerosis and osteoporosis, appear early in adulthood, and death in the fourth to sixth decade commonly ensues from myocardial infarction or cancer. In accord with the aging phenotype, cells from WS patients have a reduced replicative life span in culture. Genomic instability is observed at the cytogenetic level in the form of chromosome breaks and translocations and at the molecular level by multiple large deletions. The Werner syndrome gene (WRN) has recently been cloned. The predicted product is a 1,432-amino-acid protein whose central domain is homologous to members of the RecQ family of DNA helicases. Such homology does not necessarily mean that WRN encodes an active helicase. For example, the Saccharomyces cerevisiae RAD26 gene protein and the human transcription-repair coupling factor CSB (Cockayne syndrome 8) are highly homologous to known helicases, yet neither encodes an active helicase. Moreover, the Bloom's syndrome gene (BLM), discovered before WRN, is also homologous to the RecQ family of DNA helicases, though we still await demonstration that it encodes an active helicase. Here we report that the WS protein does indeed catalyze DNA unwinding.


Subject(s)
DNA Helicases/genetics , Point Mutation , Werner Syndrome/enzymology , Werner Syndrome/genetics , Adult , Amino Acid Sequence , Animals , Cell Line , Conserved Sequence , DNA Helicases/isolation & purification , DNA Helicases/metabolism , Humans , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spodoptera , Transfection
2.
Med Genet ; 24(4): 262-267, 2012 Dec.
Article in German | MEDLINE | ID: mdl-25309043

ABSTRACT

Werner syndrome is a segmental progeroid disorder with onset in adolescence or early adulthood. Typical symptoms contributing to patients' prematurely aged appearance include postpubertal development of short stature, cataracts, premature greying/thinning of scalp hair, scleroderma-like skin changes and regional atrophy of subcutaneous fat tissue. In addition, an increased rate and early onset of typical age-related diseases such as type 2 diabetes mellitus, osteoporosis, atherosclerosis, and various malignancies is observed. Werner syndrome is autosomal recessively inherited and caused by mutations in the Werner gene (WRN). To date, more than 70 WRN mutations have been identified. These are spread over the entire gene and typically represent loss of function mutations. WRN encodes a RecQ type helicase involved in DNA repair and the maintenance of DNA integrity, which is reflected by an increased genetic instability in patient cells. Despite the relative rarity of Werner syndrome, its analysis provides important general insights into the roles of DNA stability and integrity for the ageing process and the development of age-associated diseases.

3.
Science ; 269(5226): 973-7, 1995 Aug 18.
Article in English | MEDLINE | ID: mdl-7638622

ABSTRACT

A candidate gene for the chromosome 1 Alzheimer's disease (AD) locus was identified (STM2). The predicted amino acid sequence for STM2 is homologous to that of the recently cloned chromosome 14 AD gene (S182). A point mutation in STM2, resulting in the substitution of an isoleucine for an asparagine (N141l), was identified in affected people from Volga German AD kindreds. This N141l mutation occurs at an amino acid residue that is conserved in human S182 and in the mouse S182 homolog. The presence of missense mutations in AD subjects in two highly similar genes strongly supports the hypothesis that mutations in both are pathogenic.


Subject(s)
Alzheimer Disease/genetics , Chromosomes, Human, Pair 1/genetics , Membrane Proteins/genetics , Adult , Aged , Alzheimer Disease/ethnology , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/genetics , Female , Gene Expression , Germany/ethnology , Humans , Lod Score , Male , Membrane Proteins/chemistry , Middle Aged , Molecular Sequence Data , Mutation , Pedigree , Point Mutation , Presenilin-2
4.
Science ; 272(5259): 258-62, 1996 Apr 12.
Article in English | MEDLINE | ID: mdl-8602509

ABSTRACT

Werner's syndrome (WS) is an inherited disease with clinical symptoms resembling premature aging. Early susceptibility to a number of major age-related diseases is a key feature of this disorder. The gene responsible for WS (known as WRN) was identified by positional cloning. The predicted protein is 1432 amino acids in length and shows significant similarity to DNA helicases. Four mutations in WS patients were identified. Two of the mutations are splice-junction mutations, with the predicted result being the exclusion of exons from the final messenger RNA. One of the these mutations, which results in a frameshift and a predicted truncated protein, was found in the homozygous state in 60 percent of Japanese WS patients examined. The other two mutations are nonsense mutations. The identification of a mutated putative helicase as the gene product of the WS gene suggests that defective DNA metabolism is involved in the complex process of aging in WS patients.


Subject(s)
Chromosomes, Human, Pair 8/genetics , Cloning, Molecular , DNA Helicases/genetics , Mutation , Werner Syndrome/genetics , Aging/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Codon, Terminator , DNA/metabolism , DNA Helicases/chemistry , DNA, Complementary/genetics , Disease Susceptibility , Exodeoxyribonucleases , Exons/genetics , Female , Frameshift Mutation , Heterozygote , Homozygote , Humans , Male , Molecular Sequence Data , Neoplasms/etiology , Neoplasms/genetics , RecQ Helicases , Sequence Alignment , Werner Syndrome Helicase
5.
PLoS One ; 14(5): e0216223, 2019.
Article in English | MEDLINE | ID: mdl-31071155

ABSTRACT

Satellite telemetry is an increasingly utilized technology in wildlife research, and current devices can track individual animal movements at unprecedented spatial and temporal resolutions. However, as we enter the golden age of satellite telemetry, we need an in-depth understanding of the main technological, species-specific and environmental factors that determine the success and failure of satellite tracking devices across species and habitats. Here, we assess the relative influence of such factors on the ability of satellite telemetry units to provide the expected amount and quality of data by analyzing data from over 3,000 devices deployed on 62 terrestrial species in 167 projects worldwide. We evaluate the success rate in obtaining GPS fixes as well as in transferring these fixes to the user and we evaluate failure rates. Average fix success and data transfer rates were high and were generally better predicted by species and unit characteristics, while environmental characteristics influenced the variability of performance. However, 48% of the unit deployments ended prematurely, half of them due to technical failure. Nonetheless, this study shows that the performance of satellite telemetry applications has shown improvements over time, and based on our findings, we provide further recommendations for both users and manufacturers.


Subject(s)
Animals, Wild/physiology , Ecosystem , Environmental Monitoring , Geographic Information Systems , Spacecraft , Telemetry , Animals
7.
Mol Biol Cell ; 10(8): 2655-68, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10436020

ABSTRACT

Werner syndrome (WS) is a human progeroid syndrome characterized by the early onset of a large number of clinical features associated with the normal aging process. The complex molecular and cellular phenotypes of WS involve characteristic features of genomic instability and accelerated replicative senescence. The gene involved (WRN) was recently cloned, and its gene product (WRNp) was biochemically characterized as a helicase. Helicases play important roles in a variety of DNA transactions, including DNA replication, transcription, repair, and recombination. We have assessed the role of the WRN gene in transcription by analyzing the efficiency of basal transcription in WS lymphoblastoid cell lines that carry homozygous WRN mutations. Transcription was measured in permeabilized cells by [3H]UTP incorporation and in vitro by using a plasmid template containing the RNA polymerase II (RNA pol II)-dependent adenovirus major late promoter. With both of these approaches, we find that the transcription efficiency in different WS cell lines is reduced to 40-60% of the transcription in cells from normal individuals. This defect can be complemented by the addition of normal cell extracts to the chromatin of WS cells. Addition of purified wild-type WRNp but not mutated WRNp to the in vitro transcription assay markedly stimulates RNA pol II-dependent transcription carried out by nuclear extracts. A nonhelicase domain (a direct repeat of 27 amino acids) also appears to have a role in transcription enhancement, as revealed by a yeast hybrid-protein reporter assay. This is further supported by the lack of stimulation of transcription when mutant WRNp lacking this domain was added to the in vitro assay. We have thus used several approaches to show a role for WRNp in RNA pol II transcription, possibly as a transcriptional activator. A deficit in either global or regional transcription in WS cells may be a primary molecular defect responsible for the WS clinical phenotype.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , RNA Polymerase II/genetics , Transcription, Genetic , Amino Acid Sequence , Cell Extracts , Cell Line , Cell Membrane Permeability , Cell Nucleus/metabolism , Chromatin/genetics , DNA Helicases/isolation & purification , Exodeoxyribonucleases , Fluorescent Antibody Technique , Genetic Complementation Test , Humans , Molecular Sequence Data , Mutation , Plasmids/genetics , RNA/biosynthesis , RecQ Helicases , Repetitive Sequences, Amino Acid , Werner Syndrome/genetics , Werner Syndrome/pathology , Werner Syndrome Helicase
8.
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
9.
Nucleic Acids Res ; 28(2): 648-54, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10606667

ABSTRACT

Mutations in the chromosome 8p WRN gene cause Werner syndrome (WRN), a human autosomal recessive disease that mimics premature aging and is associated with genetic instability and an increased risk of cancer. All of the WRN mutations identified in WRN patients are predicted to truncate the WRN protein with loss of a C-terminal nuclear localization signal. However, many of these truncated proteins would retain WRN helicase and/or nuclease functional domains. We have used a combination of immune blot and immune precipitation assays to quantify WRN protein and its associated 3'-->5' helicase activity in genetically characterized WRN patient cell lines. None of the cell lines from patients harboring four different WRN mutations contained detectable WRN protein or immune-precipitable WRN helicase activity. Cell lines from WRN heterozygous individuals contained reduced amounts of both WRN protein and helicase activity. Quantitative immune blot analyses indicate that both lymphoblastoid cell lines and fibroblasts contain approximately 6 x 10(4)WRN molecules/cell. Our results indicate that most WRN mutations result in functionally equivalent null alleles, that WRN heterozygote effects may result from haploinsufficiency and that successful modeling of WRN pathogenesis in the mouse or in other model systems will require the use of WRN mutations that eliminate WRN protein expression.


Subject(s)
DNA Helicases/metabolism , Werner Syndrome/enzymology , Animals , Blotting, Western , Cell Line, Transformed , Exodeoxyribonucleases , Heterozygote , Humans , Mice , Plasmids , Precipitin Tests , RecQ Helicases , Transfection , Werner Syndrome/genetics , Werner Syndrome/pathology , Werner Syndrome Helicase
10.
Cancer Res ; 57(14): 2956-60, 1997 Jul 15.
Article in English | MEDLINE | ID: mdl-9230208

ABSTRACT

Werner syndrome (WS) is an autosomal recessive disease, the phenotype of which is a caricature of premature aging. WS cells and cell lines display several types of genetic instability, and WS patients have an increased risk of developing cancer. The WS locus (WRN) encodes a protein that shows significant sequence homology to the RecQ family of DNA helicases. Because a DNA helicase may function in DNA mismatch repair, we examined extracts of WS cell lines for mismatch repair activity. Extracts from four different WS lymphoblastoid cell lines containing different WRN mutations and from three within-pedigree control cell lines were all proficient in mismatch repair. In marked contrast, extracts from three independent WS fibroblastoid cell lines were deficient in repair of base-base and insertion/deletion mismatches. Extracts of one of these lines restored activity to extracts of mismatch repair-deficient tumor cells with defined mutations in hMSH2, hMSH3, hMSH6, hMLH1, or hPMS2. This suggests that the WRN mutation in this fibroblast line is not a dominant negative inhibitor of mismatch repair activity and that the repair defect does not reside in these five known mismatch repair genes. Defective mismatch repair in fibroblastoid but not lymphoblastoid cells is consistent with the possibility that WRN protein could have a cell type- and/or tissue-specific role in mismatch repair. Alternatively, a mutation in WRN could predispose cells to mutations in other genes required for mismatch repair activity, at least one of which could be an unknown gene.


Subject(s)
DNA Helicases/physiology , DNA Repair , Werner Syndrome/genetics , Cell Line , Exodeoxyribonucleases , Genetic Complementation Test , Humans , RecQ Helicases , Werner Syndrome Helicase
11.
Cancer Res ; 60(9): 2372-6, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10811112

ABSTRACT

WRN encodes a RecQ helicase, which is mutated in Werner syndrome. Werner syndrome is a genetic condition of young adults characterized by premature aging, limited replicative capacity of cells in vitro, and increased cancer risk. Telomerase is a reverse transcriptase that extends the G-rich strand of telomeric DNA. Primary cells in vitro typically lack telomerase activity and undergo senescence, whereas telomerase is reactivated in many, but not all, tumors. The roles of the two genes are not known to be related. Here we report the development of an effective colony-forming assay in which a SV40-transformed Werner fibroblast cell line is 6-18-fold more sensitive to 4-nitroquinoline 1-oxide than SV40-transformed normal cell lines. The sensitivity can be partially reversed by transfecting a normal WRN gene but not a mutated WRN gene into the cells. Curiously, the sensitivity can be reversed equally well by transfecting a telomerase gene (TERT) into the cells. These data indicate the possibility of an interdependent function of these two genes.


Subject(s)
4-Nitroquinoline-1-oxide/metabolism , DNA Helicases/metabolism , Mutagens/metabolism , RNA , Telomerase/metabolism , Werner Syndrome/enzymology , Animals , Blotting, Western , COS Cells , Cell Line, Transformed , DNA Helicases/genetics , DNA, Complementary/metabolism , DNA-Binding Proteins , Dose-Response Relationship, Drug , Exodeoxyribonucleases , Fibroblasts/metabolism , HeLa Cells , Humans , Models, Genetic , Molecular Sequence Data , Mutation , RecQ Helicases , Sequence Analysis, DNA , Telomerase/genetics , Telomere/genetics , Transfection , Werner Syndrome/genetics , Werner Syndrome Helicase
12.
Cancer Res ; 60(9): 2492-6, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10811130

ABSTRACT

Werner syndrome (WRN) is an uncommon autosomal recessive disease in which progeroid features are associated with genetic instability and an elevated risk of neoplasia. We have used the glycophorin A (GPA) somatic cell mutation assay to analyze genetic instability in vivo in WRN patients and heterozygotes. GPA variant frequencies were determined for 11 WRN patients and for 10 heterozygous family members who collectively carry 10 different WRN mutations. Genetic instability as measured by GPA O/N allele loss variant frequency was significantly increased, and this increase was strongly age-dependent in WRN patients. GPA O/N allele loss variants were also significantly elevated in heterozygous family members, thus providing the first evidence for in vivo genetic instability in heterozygous carriers in an autosomal recessive genetic instability syndrome. Our results and comparable data on other human genetic instability syndromes allow an estimate of the level of genetic instability that increases the risk of human bone marrow dysfunction or neoplasia.


Subject(s)
Hematologic Diseases/genetics , Heterozygote , Werner Syndrome/genetics , Adolescent , Adult , Age Factors , Aged , Alleles , Case-Control Studies , DNA Helicases/genetics , Exodeoxyribonucleases , Family Health , Female , Flow Cytometry , Genotype , Glycophorins/metabolism , Humans , Male , Middle Aged , Mutation , RecQ Helicases , Risk Factors , Werner Syndrome Helicase
13.
Oncogene ; 8(11): 2987-93, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8414501

ABSTRACT

Immortalization of rat lung epithelial cells by either wild-type SV-40 T antigen, a mutant form of T antigen that cannot bind pRb, or a temperature-sensitive T antigen increased by five- to 20-fold the steady state levels of p34cdc2 and cyclin A, positive regulators of progression through the cell cycle. Increased abundance of p34cdc2 was not accompanied by equivalent increases in cdc2 mRNA, indicating that increased expression of p34cdc2 is due, at least partially, to post-transcriptional mechanisms. Levels of p34cdc2 and cyclin A protein in cells immortalized with a temperature-sensitive T antigen remained elevated at the restrictive temperature unless T antigen was reduced to levels significantly below those where proliferation ceased, indicating that these two functions can be dissociated. These results show that SV-40 T antigen can dramatically enhance the expression of certain cell cycle regulatory proteins by mechanisms that are independent of pRb binding and cell growth status.


Subject(s)
Antigens, Polyomavirus Transforming/physiology , CDC2 Protein Kinase/analysis , Cell Transformation, Viral , Cyclins/analysis , Simian virus 40/immunology , Animals , CDC2 Protein Kinase/genetics , Cell Division , Cell Line, Transformed , Cyclins/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
14.
Genetics ; 154(1): 357-62, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10628995

ABSTRACT

Mutations at the Werner helicase locus (WRN) are responsible for the Werner syndrome (WS). WS patients prematurely develop an aged appearance and various age-related disorders. We have generated transgenic mice expressing human WRN with a putative dominant-negative mutation (K577M-WRN). Primary tail fibroblast cultures from K577M-WRN mice showed three characteristics of WS cells: hypersensitivity to 4-nitroquinoline-1-oxide (4NQO), reduced replicative potential, and reduced expression of the endogenous WRN protein. These data suggest that K577M-WRN mice may provide a novel mouse model for the WS.


Subject(s)
DNA Helicases/genetics , Genes, Dominant , Werner Syndrome/genetics , 4-Nitroquinoline-1-oxide/analogs & derivatives , 4-Nitroquinoline-1-oxide/pharmacology , Alleles , Animals , Cell Division , Down-Regulation , Exodeoxyribonucleases , Humans , Mice , Mice, Transgenic , Phenotype , Quinolones/pharmacology , RecQ Helicases , Werner Syndrome Helicase
15.
Mech Ageing Dev ; 122(11): 1121-33, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11389928

ABSTRACT

Werner syndrome (WS) is a human premature aging syndrome, which is associated with high frequencies of neoplasia and genetic instability. We have examined the occurrence of microsatellite instability, which may result from defective mismatch repair, in lymphoblastoid cell lines derived from nine WS patients. Instability was measured at the D2S123 locus by gel analysis of PCR products. Three WS cell lines had 4-13% altered alleles, compared with 0% in the other six lines. The increased frequency of microsatellite instability could not readily be associated with overt cancer or any other known clinical condition in the three patients. To examine whether the WS defect affected the humoral immune system, we measured the hypermutation of immunoglobulin variable genes in peripheral blood cells from the WS patient who donated the cell line with the highest frequency of microsatellite instability. The frequency and pattern of mutation was similar to that from normal individuals, suggesting that the Werner protein is not involved in generating hypermutation.


Subject(s)
Gene Rearrangement, B-Lymphocyte, Heavy Chain , Immunoglobulin Variable Region/genetics , Microsatellite Repeats , Mutation , Werner Syndrome/genetics , Base Sequence , Cell Line , Complementarity Determining Regions , DNA, Complementary , Humans , Molecular Sequence Data , Werner Syndrome/immunology
16.
Exp Gerontol ; 34(8): 935-41, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10673147

ABSTRACT

Werner syndrome is a human progeroid syndrome caused by mutations at the Werner helicase locus (WRN). Progeroid features and diseases associated with aging (including arteriosclerosis) do not become apparent until after puberty. We entertained two alternative hypotheses to explain the post-pubertal onset: 1) WRN expression is induced at the time of puberty, its earlier functions being satisfied by another member of that family of helicases; and 2) it is expressed at all ages, but the phenotype of deficiency becomes apparent only after puberty. We report initial experiments consistent with the second hypothesis. Steady-state levels of WRN mRNA in aortic tissues were determined by semiquantitative reverse transcription-polymerase chain reaction. WRN mRNA was detectable as early as 49 days of gestation (the earliest available material). There was no statistically significant change in these levels between fetal and adult tissues. The presence of the WRN protein in fetal aorta was confirmed by Western analysis. This rules out the possibility that Werner syndrome phenotypes manifest after the puberty because of peripubertal induction of WRN expression.


Subject(s)
Aging/metabolism , Aorta/enzymology , DNA Helicases/metabolism , Fetus/enzymology , Werner Syndrome/genetics , Werner Syndrome/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Female , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
17.
Am J Med Genet ; 68(4): 494-8, 1997 Feb 11.
Article in English | MEDLINE | ID: mdl-9021029

ABSTRACT

The Werner syndrome (WS) is a rare autosomal recessive progeroid syndrome characterized by the premature onset of multiple age-related disorders, including atherosclerosis, cancer, non-insulin-dependent diabetes mellitus (NIDDM), ocular cataracts and osteoporosis [Epstein et al., 1966]. The major cause of death (at a median age of 47) is myocardial infarction (MI) [Epstein et al., 1966]. The WS mutation involves a member (WRN) of the RecQ family of helicases and may perturb DNA replication, repair, recombination, transcription, or chromosomal segregation [Yu et al., 1996]. We now report data on 149 MI cases and age-matched controls suggesting that a polymorphic WRN variant is associated with increased risk for MI. Based on our data, homozygosity for a cysteine at amino acid 1367 (the most prevalent genotype) predicts a 2.78 times greater risk of MI (95% confidence intervals: 1.23 to 6.86). The variant was not significantly associated with NIDDM. The two alleles (cysteine vs. arginine) could influence helicase activity, turnover, macromolecular interactions or, alternatively, could be markers for haplotypes influencing WRN regulation or reflecting gene action at linked loci. However, given the caveats implicit in genetic association studies, it is imperative that the present results be replicated in independent populations.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Myocardial Infarction/genetics , Polymorphism, Genetic , Werner Syndrome/epidemiology , Werner Syndrome/genetics , Adult , Age of Onset , Aged , Arginine/genetics , Case-Control Studies , Cysteine/genetics , Diabetes Mellitus, Type 2/epidemiology , Female , Genetic Variation , Homozygote , Humans , Japan/epidemiology , Male , Middle Aged , Mutation , Myocardial Infarction/epidemiology
18.
Am J Med Genet ; 95(4): 374-80, 2000 Dec 11.
Article in English | MEDLINE | ID: mdl-11186893

ABSTRACT

Werner syndrome (WS) is a progeroid syndrome caused by autosomal recessive null mutations at the WRN locus. The WRN gene encodes a nuclear protein of 180 kD that contains both exonuclease and helicase domains. WS patients develop various forms of arteriosclerosis, particularly atherosclerosis, and medial calcinosis. The most common cause of death in Caucasian subjects with WS is myocardial infarction. Previous studies have identified specific polymorphisms within WRN that may modulate the risk of atherosclerosis. Population studies of the 1074Leu/Phe and 1367Cys/Arg polymorphisms were undertaken to evaluate the role of WRN in atherogenesis. Frequencies of the 1074Leu/Phe polymorphisms in Finnish and Mexican populations revealed an age-dependent decline of 1074Phe/Phe genotype. In Mexican newborns, but not in Finnish newborns, the 1074Leu/Phe and 1367Cys/ Arg polymorphisms were in linkage disequilibrium. Among coronary artery disease subjects, there was a tendency for the 1074Phe allele to be associated with coronary stenosis in a gene dose-dependent manner. Furthermore, the 1367Arg/Arg genotype predicted a lower degree of coronary artery occlusion, as measured by NV50, when compared to the 1367Cys/Cys or 1367Cys/Arg genotypes. However, these tendencies did not achieve statistical significance. Samples from Mexican patients with ischemic stroke showed a trend of haplotype frequencies different from that in a control group of Mexican adults. These data support the hypothesis that WRN may mediate not only WS, but may also modulate more common age-related disorders and, perhaps, a basic aging process.


Subject(s)
Amino Acid Substitution/genetics , Arteriosclerosis/genetics , Longevity/genetics , Polymorphism, Genetic/genetics , Werner Syndrome/genetics , Adult , Aged , Aged, 80 and over , Aging/genetics , Arginine/genetics , Arteriosclerosis/epidemiology , Coronary Artery Disease/epidemiology , Coronary Artery Disease/genetics , Cysteine/genetics , Finland/epidemiology , Gene Frequency , Genotype , Haplotypes , Humans , Infant, Newborn , Leucine/genetics , Mexico/epidemiology , Middle Aged , Phenylalanine/genetics , Werner Syndrome/epidemiology
19.
Am J Med Genet ; 82(5): 399-403, 1999 Feb 19.
Article in English | MEDLINE | ID: mdl-10069711

ABSTRACT

The Werner syndrome gene (WRN) encodes a novel helicase of 1,432 amino acids. Homozygous mutations, all of which result in the truncation of the protein, lead to Werner syndrome. However, little is known about the role of WRN in "normal" aging. We have identified four missense polymorphisms and four conservative polymorphsims in WRN gene. A single study showed that a polymorphism at amino acid 1367 Cys(TTG)/ Arg(CTG) is associated with a variation in risk of myocardial infarction among a Japanese population. The 1367 Cys/Arg polymorphism was examined during aging in three different populations: Finnish, Mexican, and North American. The frequencies of 1367 Cys were higher than those of 1367 Arg in all the populations examined, though the frequencies varied among populations. The frequency of the 1367 Arg allele, thought to be protective against myocardial infarction in a Japanese population, was approximately three times higher in the North American and Finnish adult populations. When newborns and centenarians were compared within the Finnish population, no differences were observed in the proportions of 1367 Cys/Arg across age groups. Within the Finnish population, we confirmed a significant decrease of the APOE epsilon2 allele and an increase in the epsilon4 allele in newborn infants compared with centenarians. Thus, unlike the APOE polymorphism, there is no evidence of an association of this WRN polymorphism with longevity.


Subject(s)
Polymorphism, Genetic , Werner Syndrome/genetics , Adult , Aged , Aged, 80 and over , Aging , Apolipoproteins E/genetics , Blood/metabolism , Fetal Blood/metabolism , Finland , Gene Frequency , Genotype , Humans , Infant, Newborn , Middle Aged , Models, Genetic , Mutation, Missense , Polymorphism, Restriction Fragment Length , Werner Syndrome/ethnology
20.
Mutat Res ; 554(1-2): 351-64, 2004 Oct 04.
Article in English | MEDLINE | ID: mdl-15450431

ABSTRACT

In mammalian cells, DNA double-strand breaks are repaired by non-homologous end-joining and homologous recombination, both pathways being essential for the maintenance of genome integrity. We determined the effect of mutations in Ku86 and DNA-PK on the efficiency and the accuracy of double-strand break repair by non-homologous end-joining and homologous recombination in mammalian cells. We used an assay, based on the transient transfection of a linearized plasmid DNA, designed to simultaneously detect transfection and recombination markers. In agreement with previous results non-homologous end-joining was largely compromised in Ku86 deficient cells, and returned to normal in the Ku86-complemented isogenic cell line. In addition, analysis of DNA plasmids recovered from Ku86 mutant cells showed an increased use of microhomologies at the nonhomologous end joining junctions, and displayed a significantly higher frequency of DNA insertions compared to control cells. On the other hand, the DNA-PKcs deficient cell lines showed efficient double-strand break repair by both mechanisms.


Subject(s)
Antigens, Nuclear/genetics , DNA-Binding Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Recombination, Genetic/genetics , Transfection , Animals , Base Sequence , CHO Cells , Cricetinae , DNA Primers , DNA-Activated Protein Kinase , Ku Autoantigen
SELECTION OF CITATIONS
SEARCH DETAIL