Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 14016, 2019 Sep 30.
Article in English | MEDLINE | ID: mdl-31570747

ABSTRACT

Werner syndrome protein (WRN) and Fanconi anemia group J protein (FANCJ) are human DNA helicases that contribute to genome maintenance. They interact with replication protein A (RPA), and these interactions dramatically enhance the unwinding activities of both helicases. Even though the interplay between these helicases and RPA is particularly important in the chemoresistance pathway of cancer cells, the precise binding regions, interfaces, and properties have not yet been characterized. Here we present systematic NMR analyses and fluorescence polarization anisotropy assays of both helicase-RPA interactions for defining core binding regions and binding affinities. Our results showed that two acidic repeats of human WRN bind to RPA70N and RPA70A. For FANCJ, the acidic-rich sequence in the C-terminal domain is the binding region for RPA70N. Our results suggest that each helicase interaction has unique features, although they both fit an acidic peptide into a basic cleft for RPA binding. Our findings shed light on the protein interactions involved in overcoming the DNA-damaging agents employed in the treatment of cancer and thus potentially provide insight into enhancing the efficacy of cancer therapy.


Subject(s)
Fanconi Anemia Complementation Group Proteins/metabolism , Fanconi Anemia/enzymology , RNA Helicases/metabolism , Werner Syndrome Helicase/metabolism , Werner Syndrome/enzymology , Binding Sites , Humans , Magnetic Resonance Spectroscopy , Replication Protein A/metabolism
2.
DNA Repair (Amst) ; 73: 17-27, 2019 01.
Article in English | MEDLINE | ID: mdl-30413344

ABSTRACT

The accelerated ageing disease Werner Syndrome (WRN) is characterized by pronounced atherosclerosis. Here, we investigated the influence of WRN downregulation on the functionality of non-replicating human endothelial cells. RNAi-mediated downregulation of WRN reduces cell motility and enhances the expression of factors regulating adhesion, inflammation, hemostasis and vasomotor tone. Moreover, WRN influences endothelial barrier function and Ca2+-release, while cell adhesion, Dil-acLDL-uptake and the mRNA expression of NO-synthases (eNOS, iNOS) remained unaffected. Regarding motility, we propose that WRN affects Rac1/FAK/ß1-integrin-related mechanisms regulating cell polarity and directed motility. Since oxidative DNA base damage contributes to aging and atherosclerosis and WRN affects DNA repair, we investigated whether downregulation of base excision repair (BER) factors mimics the effects of WRN knock-down. Indeed, downregulation of particular WRN-interacting base excision repair (BER) proteins (APE1, NEIL1, PARP1) imitates the inhibitory effect of WRN on motility. Knock-down of OGG1, which does not interact with WRN, does not influence motility but increases the mRNA expression of E-selectin, ICAM, VCAM, CCL2 and VEGFR and stimulates adhesion. Thus, individual BER factors themselves differently impact endothelial cell functionality and homeostasis. Impairment of endothelial activities caused by genotoxic stressor (tBHQ) remained largely unaffected by WRN. Summarizing, both WRN, WRN-associated BER proteins and OGG1 promote the maintenance of endothelial cell homeostasis, thereby counteracting the development of ageing-related endothelial malfunction in non-proliferating endothelial cells.


Subject(s)
DNA Helicases/metabolism , DNA Repair , Homeostasis , Human Umbilical Vein Endothelial Cells/metabolism , Werner Syndrome/enzymology , Calcium/metabolism , Cell Adhesion , Cell Movement , Gene Expression , Humans , Lipoproteins, LDL/metabolism , Protein Transport
3.
Sci Rep ; 7: 44081, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276523

ABSTRACT

Loss-of-function mutations in the WRN helicase gene cause Werner syndrome- a progeroid syndrome with an elevated risk of cancer and other age-associated diseases. Large numbers of single nucleotide polymorphisms have been identified in WRN. We report here the organismal, cellular, and molecular phenotypes of variant rs3087425 (c. 2500C > T) that results in an arginine to cysteine substitution at residue 834 (R834C) and up to 90% reduction of WRN helicase activity. This variant is present at a high (5%) frequency in Mexico, where we identified 153 heterozygous and three homozygous individuals among 3,130 genotyped subjects. Family studies of probands identified ten additional TT homozygotes. Biochemical analysis of WRN protein purified from TT lymphoblast cell lines confirmed that the R834C substitution strongly and selectively reduces WRN helicase, but not exonuclease activity. Replication track analyses showed reduced replication fork progression in some homozygous cells following DNA replication stress. Among the thirteen TT homozygotes, we identified a previously unreported and statistically significant gender bias in favor of males (p = 0.0016), but none of the clinical findings associated with Werner syndrome. Our results indicate that WRN helicase activity alone is not rate-limiting for the development of clinical WS.


Subject(s)
Homozygote , Mutation, Missense , Phenotype , Werner Syndrome Helicase/metabolism , Werner Syndrome/genetics , Adolescent , Adult , Aged , Amino Acid Substitution , Family , Female , Humans , Male , Middle Aged , Werner Syndrome/enzymology , Werner Syndrome/pathology , Werner Syndrome Helicase/genetics
4.
J Biol Chem ; 292(8): 3154-3163, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28069813

ABSTRACT

The structural and biophysical properties typically associated with G-quadruplex (G4) structures render them a significant block for DNA replication, which must be overcome for cell division to occur. The Werner syndrome protein (WRN) is a RecQ family helicase that has been implicated in the efficient processing of G4 DNA structures. The aim of this study was to identify the residues of WRN involved in the binding and ATPase-driven unwinding of G4 DNA. Using a c-Myc G4 DNA model sequence and recombinant WRN, we have determined that the RecQ-C-terminal (RQC) domain of WRN imparts a 2-fold preference for binding to G4 DNA relative to non-G4 DNA substrates. NMR studies identified residues involved specifically in interactions with G4 DNA. Three of the amino acids in the WRN RQC domain that exhibited the largest G4-specific changes in NMR signal were then mutated alone or in combination. Mutating individual residues implicated in G4 binding had a modest effect on WRN binding to DNA, decreasing the preference for G4 substrates by ∼25%. Mutating two G4-interacting residues (T1024G and T1086G) abrogated preferential binding of WRN to G4 DNA. Very modest decreases in G4 DNA-stimulated ATPase activity were observed for the mutant enzymes. Most strikingly, G4 unwinding by WRN was inhibited ∼50% for all three point mutants and >90% for the WRN double mutant (T1024G/T1086G) relative to normal B-form dsDNA substrates. Our work has helped to identify residues in the WRN RQC domain that are involved specifically in the interaction with G4 DNA.


Subject(s)
DNA/metabolism , G-Quadruplexes , Werner Syndrome Helicase/metabolism , Werner Syndrome/enzymology , DNA/chemistry , DNA/genetics , DNA Repair , DNA Replication , Humans , Models, Molecular , Mutation , Protein Domains , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase/chemistry , Werner Syndrome Helicase/genetics
5.
Dan Med J ; 63(11)2016 Nov.
Article in English | MEDLINE | ID: mdl-27808039

ABSTRACT

The growing proportion of elderly people represents an increasing economic burden, not least because of age-associated diseases that pose a significant cost to the health service. Finding possible interventions to age-associated disorders therefore have wide ranging implications. A number of genetically defined accelerated aging diseases have been characterized that can aid in our understanding of aging. Interestingly, all these diseases are associated with defects in the maintenance of our genome. A subset of these disorders, Cockayne syndrome, Xeroderma pigmentosum group A and ataxia-telangiectasia, show neurological involvement reminiscent of what is seen in primary human mitochondrial diseases. Mitochondria are the power plants of the cells converting energy stored in oxygen, sugar, fat, and protein into ATP, the energetic currency of our body. Emerging evidence has linked this organelle to aging and finding mitochondrial dysfunction in accelerated aging disorders thereby strengthens the mitochondrial theory of aging. This theory states that an accumulation of damage to the mitochondria may underlie the process of aging. Indeed, it appears that some accelerated aging disorders that show neurodegeneration also have mitochondrial dysfunction. The mitochondrial alterations may be secondary to defects in nuclear DNA repair. Indeed, nuclear DNA damage may lead to increased energy consumption, alterations in mitochondrial ATP production and defects in mitochondrial recycling, a term called mitophagy. These changes may be caused by activation of poly-ADP-ribose-polymerase 1 (PARP1), an enzyme that responds to DNA damage. Upon activation PARP1 utilizes key metabolites that attenuate pathways that are normally protective for the cell. Notably, pharmacological inhibition of PARP1 or reconstitution of the metabolites rescues the changes caused by PARP1 hyperactivation and in many cases reverse the phenotypes associated with accelerated aging. This implies that modulation of PARP1 or the downstream metabolites may be a therapeutic strategy for treating accelerated aging disorders and potentially age-associated neurological decline seen in the normal population.


Subject(s)
Aging, Premature/genetics , Aging, Premature/metabolism , Cockayne Syndrome/physiopathology , DNA Repair/genetics , Mitochondria/physiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Animals , Ataxia Telangiectasia/genetics , Bloom Syndrome/genetics , Cockayne Syndrome/genetics , DNA Repair/physiology , Dyskeratosis Congenita/genetics , Fanconi Anemia/genetics , Humans , Mitophagy , NAD/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Progeria/genetics , Progeria/metabolism , Rothmund-Thomson Syndrome/genetics , Sirtuin 1/metabolism , Telomere Shortening , Werner Syndrome/enzymology , Werner Syndrome/genetics , Xeroderma Pigmentosum/genetics
6.
Nat Commun ; 6: 8331, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26420422

ABSTRACT

Telomeric abnormalities caused by loss of function of the RecQ helicase WRN are linked to the multiple premature ageing phenotypes that characterize Werner syndrome. Here we examine WRN's role in telomeric maintenance, by comparing its action on a variety of DNA structures without or with telomeric sequences. Our results show that WRN clearly prefers to act on strand invasion intermediates in a manner that favours strand invasion and exchange. Moreover, WRN unwinding of these recombination structures is further enhanced when the invading strand contains at least three G-rich single-stranded telomeric repeats. These selectivities are most pronounced at NaCl concentrations within the reported intranuclear monovalent cation concentration range, and are partly conferred by WRN's C-terminal region. Importantly, WRN's specificity for the G-rich telomeric sequence within this precise structural context is particularly relevant to telomere metabolism and strongly suggests a physiological role in telomeric recombination processes, including T-loop dynamics.


Subject(s)
DNA/chemistry , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/genetics , RecQ Helicases/chemistry , RecQ Helicases/genetics , Recombination, Genetic , Telomere/metabolism , Werner Syndrome/enzymology , DNA/genetics , DNA/metabolism , Humans , Telomere/genetics , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase
7.
PLoS One ; 10(10): e0140292, 2015.
Article in English | MEDLINE | ID: mdl-26447695

ABSTRACT

Werner syndrome (WS) is a premature aging disorder caused by mutations in a RecQ-family DNA helicase, WRN. Mice lacking part of the helicase domain of the WRN orthologue exhibit many phenotypic features of WS, including metabolic abnormalities and a shorter mean life span. In contrast, mice lacking the entire Wrn protein (i.e. Wrn null mice) do not exhibit a premature aging phenotype. In this study, we used a targeted mass spectrometry-based metabolomic approach to identify serum metabolites that are differentially altered in young Wrn helicase mutant and Wrn null mice. An antibody-based quantification of 43 serum cytokines and markers of cardiovascular disease risk complemented this study. We found that Wrn helicase mutants exhibited elevated and decreased levels, respectively, of the anti-inflammatory cytokine IL-10 and the pro-inflammatory cytokine IL-18. Wrn helicase mutants also exhibited an increase in serum hydroxyproline and plasminogen activator inhibitor-1, markers of extracellular matrix remodeling of the vascular system and inflammation in aging. We also observed an abnormal increase in the ratio of very long chain to short chain lysophosphatidylcholines in the Wrn helicase mutants underlying a peroxisome perturbation in these mice. Remarkably, the Wrn mutant helicase protein was mislocalized to the endoplasmic reticulum and the peroxisomal fractions in liver tissues. Additional analyses with mouse embryonic fibroblasts indicated a severe defect of the autophagy flux in cells derived from Wrn helicase mutants compared to wild type and Wrn null animals. These results indicate that the deleterious effects of the helicase-deficient Wrn protein are mediated by the dysfunction of several cellular organelles.


Subject(s)
RecQ Helicases/genetics , Werner Syndrome/genetics , Animals , Autophagy , Cells, Cultured , Endoplasmic Reticulum/enzymology , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Oxidative Stress , Phenotype , Protein Transport , Reactive Oxygen Species/metabolism , Werner Syndrome/blood , Werner Syndrome/enzymology , Werner Syndrome Helicase
8.
BMC Med Genet ; 16: 91, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26449372

ABSTRACT

BACKGROUND: Putative G-quadruplex-forming sequences (PQS) have long been implicated in regulation of transcription, though the actual mechanisms are not well understood. One proposed mechanism involves the activity of PQS-specific helicases belonging to the RecQ helicase family. However, patterns of PQS that correlate with transcriptional sensitivity to RecQ helicases are not well studied, and no adequate transcriptional model exists to account for PQS effects. METHODS: To better understand PQS transcriptional effects, we analyze PQS motifs in genes differentially-transcribed in Bloom Syndrome (BS) and Werner Syndrome (WS), two disorders resulting in loss of PQS-interacting RecQ helicases.  We also correlate PQS genome-wide with transcription in multiple human cells lines while controlling for epigenetic status.  Finally, we perform neural network clustering of PQS motifs to assess whether certain motifs are over-represented in genes sensitive to RecQ helicase loss. RESULTS: By analyzing PQS motifs in promoters of genes differentially-transcribed in BS and WS, we demonstrate that abundance of promoter PQS is generally higher in down-regulated genes and lower in up-regulated genes, and show that these effects are position-dependent. To interpret these correlations we determined genome-wide PQS correlations with transcription while controlling for epigenetic status. Our results identify multiple discrete transcription start site-proximal positions where PQS are correlated with either increased or decreased transcription. Finally, we report neural network clustering analysis of PQS motifs demonstrating that genes differentially-expressed in BS and WS are significantly biased in PQS motif composition. CONCLUSIONS: Our findings unveil unappreciated detail in the relationship between PQS, RecQ helicases, and transcription. We show that promoter PQS are generally correlated with reduced gene expression, and that this effect is relieved by RecQ helicases. We also show that PQS at certain positions on the downstream sense strand are correlated with increased transcription. We therefore propose a new transcriptional model in which promoter PQS have at least two distinct types of transcriptional regulatory effects.


Subject(s)
Bloom Syndrome/genetics , DNA/chemistry , G-Quadruplexes , RecQ Helicases/metabolism , Transcription, Genetic , Werner Syndrome/genetics , Bloom Syndrome/enzymology , Cell Line , Computational Biology/methods , Epigenesis, Genetic , Gene Expression Regulation , Genome, Human , Humans , Models, Genetic , Promoter Regions, Genetic , Werner Syndrome/enzymology
9.
Biogerontology ; 14(1): 47-62, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23112078

ABSTRACT

Werner Syndrome (WS) is a human segmental progeria resulting from mutations in a DNA helicase. WS fibroblasts have a shortened replicative capacity, an aged appearance, and activated p38 MAPK, features that can be modulated by inhibition of the p38 pathway. Loss of the WRNp RecQ helicase has been shown to result in replicative stress, suggesting that a link between faulty DNA repair and stress-induced premature cellular senescence may lead to premature ageing in WS. Other progeroid syndromes that share overlapping pathophysiological features with WS also show defects in DNA processing, raising the possibility that faulty DNA repair, leading to replicative stress and premature cellular senescence, might be a more widespread feature of premature ageing syndromes. We therefore analysed replicative capacity, cellular morphology and p38 activation, and the effects of p38 inhibition, in fibroblasts from a range of progeroid syndromes. In general, populations of young fibroblasts from non-WS progeroid syndromes do not have a high level of cells with an enlarged morphology and F-actin stress fibres, unlike young WS cells, although this varies between strains. p38 activation and phosphorylated HSP27 levels generally correlate well with cellular morphology, and treatment with the p38 inhibitor SB203580 effects cellular morphology only in strains with enlarged cells and phosphorylated HSP27. For some syndromes fibroblast replicative capacity was within the normal range, whereas for others it was significantly shorter (e.g. HGPS and DKC). However, although in most cases SB203580 extended replicative capacity, with the exception of WS and DKC the magnitude of the effect was not significantly different from normal dermal fibroblasts. This suggests that stress-induced premature cellular senescence via p38 activation is restricted to a small subset of progeroid syndromes.


Subject(s)
Cellular Senescence/physiology , Werner Syndrome/enzymology , Werner Syndrome/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Ataxia Telangiectasia/enzymology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Genomic Instability , Humans , Imidazoles/pharmacology , Progeria/enzymology , Progeria/genetics , Progeria/pathology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Stress, Physiological , Syndrome , Telomerase/metabolism , Werner Syndrome/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
10.
Cell Cycle ; 11(16): 3036-44, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22871734

ABSTRACT

Werner syndrome (WS) is a disorder characterized by features of premature aging and increased cancer that is caused by loss of the RecQ helicase WRN. Telomeres consisting of duplex TTAGGG repeats in humans protect chromosome ends and sustain cellular proliferation. WRN prevents the loss of telomeres replicated from the G-rich strand, which can form secondary G-quadruplex (G4) structures. Here, we dissected WRN roles in the replication of telomeric sequences by examining factors inherent to telomeric repeats, such as G4 DNA, independently from other factors at chromosome ends that can also impede replication. For this we used the supF shuttle vector (SV) mutagenesis assay. We demonstrate that SVs with [TTAGGG]6 sequences are stably replicated in human cells, and that the repeats suppress the frequency of large deletions despite G4 folding potential. WRN depletion increased the supF mutant frequency for both the telomeric and non-telomeric SVs, compared with the control cells, but this increase was much greater (27-fold) for telomeric SVs. The higher SV mutant frequencies in WRN-deficient cells were primarily due to an increase in large sequence deletions and rearrangements. However, WRN depletion caused a more dramatic increase in deletions and rearrangements arising within the telomeric SV (70-fold), compared with non-telomeric SV (8-fold). Our results indicate that WRN prevents large deletions and rearrangements during replication, and that this role is particularly important in templates with telomeric sequence. This provides a possible explanation for increased telomere loss in WS cells.


Subject(s)
DNA Replication , Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Sequence Deletion , Telomere/metabolism , Base Sequence , Cell Line, Tumor , Exodeoxyribonucleases/genetics , G-Quadruplexes , Gene Rearrangement , Genes, Reporter , Genes, Suppressor , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Molecular Sequence Data , Mutagenesis , RNA, Transfer/genetics , RNA, Transfer/metabolism , RecQ Helicases/genetics , Repetitive Sequences, Nucleic Acid , Telomere/genetics , Werner Syndrome/enzymology , Werner Syndrome/genetics , Werner Syndrome Helicase
13.
Biochem Biophys Res Commun ; 411(4): 684-9, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21763283

ABSTRACT

Werner syndrome (WS) is an autosomal recessive premature aging disorder characterized by aging-related phenotypes and genomic instability. WS is caused by mutations in a gene encoding a nuclear protein, Werner syndrome protein (WRN), a member of the RecQ helicase family, that interestingly possesses both helicase and exonuclease activities. Previous studies have shown that the two activities act in concert on a single substrate. We investigated the effect of a DNA secondary structure on the two WRN activities and found that a DNA secondary structure of the displaced strand during unwinding stimulates WRN helicase without coordinate action of WRN exonuclease. These results imply that WRN helicase and exonuclease activities can act independently, and we propose that the uncoordinated action may be relevant to the in vivo activity of WRN.


Subject(s)
DNA/chemistry , Exodeoxyribonucleases/chemistry , Nucleic Acid Conformation , RecQ Helicases/chemistry , Werner Syndrome/enzymology , Humans , Oligonucleotides/chemistry , Substrate Specificity , Werner Syndrome Helicase
14.
Cell Cycle ; 10(12): 1998-2007, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21558802

ABSTRACT

XPG is a structure-specific endonuclease required for nucleotide excision repair (NER). XPG incision defects result in the cancer-prone syndrome xeroderma pigmentosum, whereas truncating mutations of XPG cause the severe postnatal progeroid developmental disorder Cockayne syndrome. We show that XPG interacts directly with WRN protein, which is defective in the premature aging disorder Werner syndrome, and that the two proteins undergo similar subnuclear redistribution in S phase and colocalize in nuclear foci. The co-localization was observed in mid- to late S phase, when WRN moves from nucleoli to nuclear foci that have been shown to contain both protein markers of stalled replication forks and telomeric proteins. We mapped the interaction between XPG and WRN to the C-terminal domains of each, and show that interaction with the C-terminal domain of XPG strongly stimulates WRN helicase activity. WRN also possesses a competing DNA single-strand annealing activity that, combined with unwinding, has been shown to coordinate regression of model replication forks to form Holliday junction/chicken foot intermediate structures. We tested whether XPG stimulated WRN annealing activity, and found that XPG itself has intrinsic strand annealing activity that requires the unstructured R- and C-terminal domains but not the conserved catalytic core or endonuclease activity. Annealing by XPG is cooperative, rather than additive, with WRN annealing. Taken together, our results suggest a novel function for XPG in S phase that is, at least in part, performed coordinately with WRN, and which may contribute to the severity of the phenotypes that occur upon loss of XPG.


Subject(s)
DNA-Binding Proteins/metabolism , Endonucleases/metabolism , Exodeoxyribonucleases/metabolism , Nuclear Proteins/metabolism , RecQ Helicases/metabolism , Transcription Factors/metabolism , Werner Syndrome/enzymology , Binding Sites , DNA Helicases , DNA Repair , DNA Replication , DNA-Binding Proteins/physiology , Endonucleases/physiology , Exodeoxyribonucleases/physiology , Humans , Nuclear Proteins/physiology , Protein Binding , RecQ Helicases/physiology , S Phase , Transcription Factors/physiology , Werner Syndrome Helicase , Xeroderma Pigmentosum
15.
Structure ; 18(2): 149-51, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20159459

ABSTRACT

In this issue of Structure, Kitano et al. describe the structure of the DNA-bound winged-helix domain from the Werner helicase. This structure of a RecQ/DNA complex offers insights into the DNA-unwinding mechanisms of RecQ family helicases.


Subject(s)
DNA , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , RecQ Helicases/chemistry , RecQ Helicases/metabolism , Werner Syndrome/enzymology , DNA/chemistry , DNA/metabolism , Exodeoxyribonucleases/genetics , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Protein Structure, Tertiary , RecQ Helicases/genetics , Werner Syndrome Helicase
16.
DNA Repair (Amst) ; 9(3): 331-44, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20075015

ABSTRACT

Werner syndrome protein (WRN) is one of a family of five human RecQ helicases implicated in the maintenance of genome stability. The conserved RecQ family also includes RecQ1, Bloom syndrome protein (BLM), RecQ4, and RecQ5 in humans, as well as Sgs1 in Saccharomyces cerevisiae, Rqh1 in Schizosaccharomyces pombe, and homologs in Caenorhabditis elegans, Xenopus laevis, and Drosophila melanogaster. Defects in three of the RecQ helicases, RecQ4, BLM, and WRN, cause human pathologies linked with cancer predisposition and premature aging. Mutations in the WRN gene are the causative factor of Werner syndrome (WS). WRN is one of the best characterized of the RecQ helicases and is known to have roles in DNA replication and repair, transcription, and telomere maintenance. Studies both in vitro and in vivo indicate that the roles of WRN in a variety of DNA processes are mediated by post-translational modifications, as well as several important protein-protein interactions. In this work, we will summarize some of the early studies on the cellular roles of WRN and highlight the recent findings that shed some light on the link between the protein with its cellular functions and the disease pathology.


Subject(s)
DNA Repair , DNA/metabolism , Genome , RecQ Helicases/metabolism , Animals , DNA Replication , Humans , RecQ Helicases/genetics , Werner Syndrome/enzymology , Werner Syndrome/genetics
17.
Biochem Pharmacol ; 79(3): 463-70, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19737542

ABSTRACT

Werner's syndrome (WS) is a rare human autosomal recessive segmental progeroid syndrome clinically characterized by atherosclerosis, cancer, osteoporosis, type 2 diabetes mellitus and ocular cataracts. The WRN gene codes for a RecQ helicase which is present in many tissues. Although the exact functions of the WRN protein remain unclear, accumulating evidence suggests that it participates in DNA repair, replication, recombination and telomere maintenance. It has also been proposed that WRN participates in RNA polymerase II-dependent transcription. However no promoter directly targeted by WRN has yet been identified. In this work, we report mammalian genes that are WRN targets. The rat CYP2B2 gene and its closely related mouse homolog, Cyp2b10, are both strongly induced in liver by phenobarbital. We found that there is phenobarbital-dependent recruitment of WRN to the promoter of the CYP2B2 gene as demonstrated by chromatin immunoprecipitation analysis. Mice homozygous for a Wrn mutation deleting part of the helicase domain showed a decrease in basal and phenobarbital-induced CYP2B10 mRNA levels compared to wild type animals. The phenobarbital-induced level of CYP2B10 protein was also reduced in the mutant mice. Electrophoretic mobility shift assays showed that WRN can participate in the formation of a complex with a specific sequence within the CYP2B2 basal promoter. Hence, there is a WRN binding site in a region of DNA sequence to which WRN is recruited in vivo. Taken together, these results suggest that WRN participates in transcription of CYP2B genes in liver and identifies the first physical interaction between a specific promoter sequence and WRN.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Liver/enzymology , Phenobarbital/pharmacology , RecQ Helicases/genetics , Steroid Hydroxylases/genetics , Transcriptional Activation/genetics , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Aryl Hydrocarbon Hydroxylases/metabolism , Chromatin/genetics , Chromatin/metabolism , Cytochrome P-450 CYP3A/biosynthesis , Cytochrome P-450 CYP3A/genetics , Cytochrome P450 Family 2 , Drug Delivery Systems , Enzyme Induction/drug effects , Enzyme Induction/genetics , Gene Deletion , Liver/drug effects , Liver/pathology , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/drug effects , Protein Transport/genetics , Rats , Rats, Sprague-Dawley , RecQ Helicases/biosynthesis , RecQ Helicases/deficiency , Steroid Hydroxylases/biosynthesis , Steroid Hydroxylases/metabolism , Transcriptional Activation/drug effects , Werner Syndrome/enzymology , Werner Syndrome/genetics
18.
DNA Repair (Amst) ; 9(1): 11-22, 2010 Jan 02.
Article in English | MEDLINE | ID: mdl-19896421

ABSTRACT

Werner syndrome (WS) is a human autosomal recessive genetic instability and cancer predisposition syndrome with features of premature aging. Several genetically determined mouse models of WS have been generated, however, none develops features of premature aging or an elevated risk of neoplasia unless additional genetic perturbations are introduced. In order to determine whether differences in cellular phenotype could explain the discrepant phenotypes of Wrn-/- mice and WRN-deficient humans, we compared the cellular phenotype of newly derived Wrn-/- mouse primary fibroblasts with previous analyses of primary and transformed fibroblasts from WS patients and with newly derived, WRN-depleted human primary fibroblasts. These analyses confirmed previously reported cellular phenotypes of WRN-mutant and WRN-deficient human fibroblasts, and demonstrated that the human WRN-deficient cellular phenotype can be detected in cells grown in 5% or in 20% oxygen. In contrast, we did not identify prominent cellular phenotypes present in WRN-deficient human cells in Wrn-/- mouse fibroblasts. Our results indicate that human and mouse fibroblasts have different functional requirements for WRN protein, and that the absence of a strong cellular phenotype may in part explain the failure of Wrn-/- mice to develop an organismal phenotype resembling Werner syndrome.


Subject(s)
Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Werner Syndrome/enzymology , Animals , Cell Proliferation , Cells, Cultured , DNA Damage , Exodeoxyribonucleases/deficiency , Histones/metabolism , Humans , Longevity , Mice , Mice, Knockout , Neoplasms/enzymology , Neoplasms/genetics , Oxygen/metabolism , Phenotype , RecQ Helicases/deficiency , Werner Syndrome/genetics , Werner Syndrome/pathology , Werner Syndrome Helicase
19.
Cell Cycle ; 8(13): 2080-92, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19502800

ABSTRACT

Werner syndrome (WS) is characterized by the premature onset of several age-associated pathologies. The protein deficient in WS (WRN) is a RecQ-type DNA helicase involved in DNA repair, replication, telomere maintenance and transcription. However, precisely how WRN deficiency leads to the numerous WS pathologies is still unknown. Here we use short-term siRNA-based inhibition of WRN to test the direct consequences of its loss on gene expression. Importantly, this short-term knock down of WRN protein level was sufficient to trigger an expression profile resembling fibroblasts established from old donor patients. In addition, this treatment altered sets of genes involved in 14 distinct biological pathways. Besides the already known impact of WRN on DNA replication, DNA repair, the p21/p53 pathway, and cell cycle, gene set enrichment analyses of our microarray data also uncover significant impact on the MYC, E2F, cellular E2A and ETV5 transcription factor pathways as well as adipocyte differentiation, HIF1, NFkappaB and IL-6 pathways. Finally, short-term siRNA-based inhibition of mouse Wrn expression in the pre-adipocyte cell line 3T3-L1 confirmed the impact of WRN on adipogenesis. These results are consistent with the pro-inflammatory status and lipid abnormalities observed in WS patients. This approach thus identified new effectors of WRN activity that might contribute to the WS phenotype.


Subject(s)
Adipogenesis/genetics , Cell Cycle/genetics , DNA Damage , Exodeoxyribonucleases/metabolism , RecQ Helicases/metabolism , Werner Syndrome/enzymology , 3T3-L1 Cells , Animals , Cells, Cultured , DNA Repair , Exodeoxyribonucleases/deficiency , Exodeoxyribonucleases/genetics , Fibroblasts/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Inflammation/genetics , Mice , RNA, Small Interfering/metabolism , RecQ Helicases/deficiency , RecQ Helicases/genetics , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase
20.
FEBS Lett ; 583(8): 1331-6, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19306876

ABSTRACT

Caloric restriction (CR) is known to effectively elongate mammalian life-spans. The compound 2-deoxy-D-glucose (2DG), which is often used as an inhibitor of glucose utilization, is a mimetic agent of CR. In this study, we examined the changes of telomerase and Werner's syndrome RecQ (WRN) helicase after treatment with 2DG, because of the involvement of recQ helicase in the regulation of telomeres. Interestingly, 2DG treatment increased the expression of WRN protein in accordance with induction of its promoter activity and gene expression. Furthermore, the activation of telomerase was observed after 2DG treatment, whereas it resulted in the reduction of cell proliferation. These results suggest that 2DG could up-regulate telomere maintenance factors accompanied with suppression of proliferation.


Subject(s)
Deoxyglucose/pharmacology , RecQ Helicases/genetics , Werner Syndrome/enzymology , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Enzyme Activation , Humans , Polymerase Chain Reaction , Promoter Regions, Genetic , Telomerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...