Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 249
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Biochem ; 86: 461-484, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28654322

ABSTRACT

Self-catalyzed DNA depurination is a sequence-specific physiological mechanism mediated by spontaneous extrusion of a stem-loop catalytic intermediate. Hydrolysis of the 5'G residue of the 5'GA/TGG loop and of the first 5'A residue of the 5'GAGA loop, together with particular first stem base pairs, specifies their hydrolysis without involving protein, cofactor, or cation. As such, this mechanism is the only known DNA catalytic activity exploited by nature. The consensus sequences for self-depurination of such G- and A-loop residues occur in all genomes examined across the phyla, averaging one site every 2,000-4,000 base pairs. Because apurinic sites are subject to error-prone repair, leading to substitution and short frameshift mutations, they are both a source of genome damage and a means for creating sequence diversity. Their marked overrepresentation in genomes, and largely unchanging density from the lowest to the highest organisms, indicate their selection over the course of evolution. The mutagenicity at such sites in many human genes is associated with loss of function of key proteins responsible for diverse diseases.


Subject(s)
Adenine/metabolism , Bloom Syndrome/genetics , DNA, Catalytic/genetics , Guanine/metabolism , Polymorphism, Genetic , Werner Syndrome/genetics , Biological Evolution , Bloom Syndrome/metabolism , Bloom Syndrome/pathology , Catalysis , DNA Repair , DNA, Catalytic/metabolism , DNA, Cruciform/genetics , DNA, Cruciform/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Humans , Hydrolysis , Inverted Repeat Sequences , Mutation , Werner Syndrome/metabolism , Werner Syndrome/pathology , beta-Globins/genetics , beta-Globins/metabolism
2.
Bioessays ; 44(8): e2200057, 2022 08.
Article in English | MEDLINE | ID: mdl-35751457

ABSTRACT

Hereditary breast and ovarian cancers are frequently attributed to germline mutations in the tumor suppressor genes BRCA1 and BRCA2. BRCA1/2 act to repair double-strand breaks (DSBs) and suppress the demise of unstable replication forks. Our work elucidated a dynamic interplay between BRCA2 and the WRN DNA helicase/exonuclease defective in the premature aging disorder Werner syndrome. WRN and BRCA2 participate in complementary pathways to stabilize replication forks in cancer cells, allowing them to proliferate. Whether the functional overlap of WRN and BRCA2 is relevant to replication at gaps between newly synthesized DNA fragments, protection of telomeres, and/or metabolism of secondary DNA structures remain to be determined. Advances in understanding the mechanisms elicited during replication stress have prompted the community to reconsider avenues for cancer therapy. Insights from studies of PARP or topoisomerase inhibitors provide working models for the investigation of WRN's mechanism of action. We discuss these topics, focusing on the implications of the WRN-BRCA2 genetic interaction under conditions of replication stress.


Subject(s)
Aging, Premature , DNA Replication , Neoplasms , Werner Syndrome , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Chromosomal Instability , DNA Helicases/chemistry , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase/genetics , Werner Syndrome Helicase/metabolism
3.
Int J Mol Sci ; 25(15)2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39125869

ABSTRACT

Werner syndrome (WS) is an autosomal recessive disease caused by loss of function of WRN. WS is a segmental progeroid disease and shows early onset or increased frequency of many characteristics of normal aging. WRN possesses helicase, annealing, strand exchange, and exonuclease activities and acts on a variety of DNA substrates, even complex replication and recombination intermediates. Here, we review the genetics, biochemistry, and probably physiological functions of the WRN protein. Although its precise role is unclear, evidence suggests WRN plays a role in pathways that respond to replication stress and maintain genome stability particularly in telomeric regions.


Subject(s)
DNA Replication , Genomic Instability , Werner Syndrome Helicase , Werner Syndrome , Werner Syndrome Helicase/metabolism , Werner Syndrome Helicase/genetics , Humans , Werner Syndrome/genetics , Werner Syndrome/metabolism , Animals , Telomere/metabolism , Telomere/genetics
4.
Am J Med Genet A ; 191(1): 220-227, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36214313

ABSTRACT

Interstitial lung disease (ILD) is a condition affecting the lung parenchyma by inflammation and fibrosis and can be caused by various exposures, connective tissue diseases (CTD), and genetic disorders. In this report, a family with five patients having progressive respiratory failure that begins with coughing in adolescence, followed by dyspnea and recurrent spontaneous pneumothorax, and death in early adulthood is presented. The patients were diagnosed to have ILD through clinical and radiological evaluations. Molecular genetic analyses of the family provided two homozygous rare variants in the WRN and SFXN5 genes, co-segregating with the phenotype. The network analyses pointed out that the variant in the WRN, rather than that in the SFXN5 gene, could be the main factor in the existence of the ILD phenotype, putatively through the altered DNA repair and telomere maintenance pathways. In silico analyses suggested that the variant could affect the exonuclease activity or the stability of the WRN protein. Moreover, the adolescent-onset pulmonary phenotype described in the case has not been reported in Werner Syndrome, the only disease known to be associated with biallelic WRN pathogenic variants. Thus, the present phenotype could be either a very atypical presentation of Werner syndrome or a new clinical entity associated with the WRN gene.


Subject(s)
Lung Diseases, Interstitial , Pneumothorax , Werner Syndrome , Humans , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/genetics , Pneumothorax/diagnosis , Pneumothorax/genetics , RecQ Helicases/genetics , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome/pathology , Werner Syndrome Helicase/genetics , Werner Syndrome Helicase/metabolism
5.
Am J Med Genet A ; 188(5): 1630-1634, 2022 05.
Article in English | MEDLINE | ID: mdl-35037378

ABSTRACT

Werner syndrome (WS) is an extremely rare, autosomal recessive segmental progeroid disorder caused by biallelic pathogenic variants in the WRN, which encodes a multifunctional nuclear protein that belongs to the RecQ family of DNA helicases. Despite extensive research on WS in the last years, the population-specific mutational spectrum still needs to be elucidated. Moreover, there is an evident lack of detailed clinical descriptions accompanied with photographs of affected individuals. Here, we report a consanguineous Lebanese family in whom we identified a pathogenic homozygous nonsense variant c.1111G>T, p.Glu371* in the WRN. The index individual, at the age of 54 years, was suspected to have WS due to a history of early-onset cataracts, premature hair loss and graying, chronic nonhealing leg ulcers, Achilles' tendon calcifications, type 2 diabetes mellitus, dyslipidemia, hypothyroidism, and premature coronary artery disease. His four sisters, three of which deceased in the fifth decade, had clinical signs suggestive of WS. Moreover, his daughter, aged 23 years, had short stature, hair loss and flat feet. Taken together, we report a detailed clinical course of disease in several affected members of a consanguineous family, which is additionally documented by photographs.


Subject(s)
Diabetes Mellitus, Type 2 , Werner Syndrome , Alopecia , Female , Humans , Male , Middle Aged , RecQ Helicases/genetics , Werner Syndrome/diagnosis , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase/genetics , Werner Syndrome Helicase/metabolism , Young Adult
6.
Cytogenet Genome Res ; 161(6-7): 297-304, 2021.
Article in English | MEDLINE | ID: mdl-34433164

ABSTRACT

Werner syndrome (WS) is an accelerated ageing disease caused by multiple mutations in the gene encoding the Werner DNA helicase (WRN). The major clinical features of WS include wrinkles, grey hair, osteoporosis, and metabolic phenomena such as atherosclerosis, diabetes, and fatty liver, and resemble those seen in normal ageing, but occur earlier, in middle age. Defective DNA repair resulting from mutations in WRN explain the majority of the clinical features of WS, but the underlying mechanisms driving the larger metabolic dysfunction remain elusive. Recent studies in animal models of WS and in WS patient cells and blood samples suggest the involvement of impaired mitophagy, NAD+ depletion, and accumulation of damaged mitochondria in metabolic dysfunction. This mini-review summarizes recent progress in the understanding of the molecular mechanisms of metabolic dysfunction in WS, with the involvement of DNA damage, mitochondrial dysfunction, mitophagy reduction, stem cell impairment, and senescence. Future studies on NAD+ and mitophagy may shed light on potential therapeutic strategies for the WS patients.


Subject(s)
Aging/genetics , DNA Damage , Mitochondria/genetics , Mitophagy/genetics , Stem Cells/metabolism , Werner Syndrome/genetics , Animals , Cellular Senescence/genetics , Humans , Mitochondria/metabolism , Telomere/genetics , Telomere/metabolism , Werner Syndrome/metabolism , Werner Syndrome/pathology
7.
Mutagenesis ; 36(5): 349-357, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34272950

ABSTRACT

G:C sites distant from 8-oxo-7,8-dihydroguanine (GO, 8-hydroxyguanine) are frequently mutated when the lesion-bearing plasmid DNA is replicated in human cells with reduced Werner syndrome (WRN) protein. To detect the untargeted mutations preferentially, the oxidised guanine base was placed downstream of the reporter supF gene and the plasmid DNA was introduced into WRN-knockdown cells. The total mutant frequency seemed higher in the WRN-knockdown cells as compared to the control cells. Mutation analyses revealed that substitution mutations occurred at the G:C pairs of 5'-GpA-3'/5'-TpC-3' sites, the preferred sequence for the apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like 3 (APOBEC3)-family cytosine deaminases, in the supF gene in both control and knockdown cells. These mutations were observed more frequently at G sites than C sites on the DNA strand where the GO base was originally located. This tendency was promoted by the knockdown of the WRN protein. The present results imply the possible involvement of APOBEC3-family cytosine deaminases in the action-at-a-distance (untargeted) mutations at G:C (or G) sites induced by GO and in cancer initiation by oxidative stress.


Subject(s)
Guanine , Mutation , Werner Syndrome Helicase/genetics , Werner Syndrome/genetics , Base Sequence , Cell Line , Gene Knockdown Techniques , Gene Order , Guanine/metabolism , Humans , Mutation Rate , Plasmids/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase/metabolism
8.
Endocr J ; 68(3): 261-267, 2021 Mar 28.
Article in English | MEDLINE | ID: mdl-33087645

ABSTRACT

Werner syndrome, also called adult progeria, is a heritable autosomal recessive human disorder characterized by the premature onset of numerous age-related diseases including juvenile cataracts, dyslipidemia, diabetes mellitus (DM), osteoporosis, atherosclerosis, and cancer. Werner syndrome is a segmental progeroid syndrome whose presentation resembles accelerated aging. The most common causes of death for WS patients are atherosclerosis and cancer. A 40-year-old female presented with short stature, bird-like facies, canities with alopecia, scleroderma-like skin changes, and non-healing foot ulcers. The patient reported a history of delayed puberty, abortion, hypertriglyceridemia, and juvenile cataracts. A clinical diagnosis of WS was made and subsequently confirmed. We discovered two WRN gene mutations in the patient, Variant 1 was the most common WRN mutation, nonsense mutation (c.1105C>T:p.R369Ter) in exon 9, which caused a premature termination codon (PTC) at position 369. Variant 2 was a frameshift mutation (c.1134delA:p.E379KfsTer5) in exon 9, which caused a PTC at position 383 and has no published reports describing. Patients with WS can show a wide variety of clinical and biological manifestations in endocrine-metabolic systems (DM, thyroid dysfunction, and hyperlipidemia). Doctors must be cognizant of early manifestations of WS and treatment options.


Subject(s)
Bone Diseases, Metabolic/physiopathology , Diabetes Mellitus, Type 2/metabolism , Fatty Liver/physiopathology , Hypertriglyceridemia/metabolism , Hypothyroidism/metabolism , Werner Syndrome/metabolism , Abortion, Habitual/physiopathology , Adipose Tissue/diagnostic imaging , Adult , Alopecia/physiopathology , Body Composition , Bone Diseases, Metabolic/diagnostic imaging , Cataract/physiopathology , Codon, Nonsense , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/physiopathology , Diabetic Foot/etiology , Diabetic Foot/physiopathology , Fatty Liver/diagnostic imaging , Female , Frameshift Mutation , Humans , Hypothyroidism/physiopathology , Intra-Abdominal Fat/diagnostic imaging , Uterus/abnormalities , Werner Syndrome/diagnosis , Werner Syndrome/genetics , Werner Syndrome/physiopathology , Werner Syndrome Helicase/genetics
9.
Nucleic Acids Res ; 47(13): 6796-6810, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31114910

ABSTRACT

Stabilization of stalled replication forks prevents excessive fork reversal or degradation, which can undermine genome integrity. The WRN protein is unique among the other human RecQ family members to possess exonuclease activity. However, the biological role of the WRN exonuclease is poorly defined. Recently, the WRN exonuclease has been linked to protection of stalled forks from degradation. Alternative processing of perturbed forks has been associated to chemoresistance of BRCA-deficient cancer cells. Thus, we used WRN exonuclease-deficiency as a model to investigate the fate of perturbed forks undergoing degradation, but in a BRCA wild-type condition. We find that, upon treatment with clinically-relevant nanomolar doses of the Topoisomerase I inhibitor camptothecin, loss of WRN exonuclease stimulates fork inactivation and accumulation of parental gaps, which engages RAD51. Such mechanism affects reinforcement of CHK1 phosphorylation and causes persistence of RAD51 during recovery from treatment. Notably, in WRN exonuclease-deficient cells, persistence of RAD51 correlates with elevated mitotic phosphorylation of MUS81 at Ser87, which is essential to prevent excessive mitotic abnormalities. Altogether, these findings indicate that aberrant fork degradation, in the presence of a wild-type RAD51 axis, stimulates RAD51-mediated post-replicative repair and engagement of the MUS81 complex to limit genome instability and cell death.


Subject(s)
Camptothecin/pharmacology , DNA Replication/drug effects , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/physiology , Endonucleases/physiology , Nucleic Acid Conformation/drug effects , Rad51 Recombinase/physiology , Topoisomerase I Inhibitors/pharmacology , Werner Syndrome Helicase/deficiency , BRCA2 Protein/physiology , Cell Line, Transformed , Checkpoint Kinase 1/metabolism , DNA Breaks, Double-Stranded , Enzyme Activation , Fibroblasts , Humans , Mitochondria/drug effects , Mitosis/drug effects , Multiprotein Complexes/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Werner Syndrome/metabolism , Werner Syndrome Helicase/physiology
10.
FASEB J ; 32(7): 3623-3640, 2018 07.
Article in English | MEDLINE | ID: mdl-29452565

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 ortholog exhibit several phenotypic features of WS. In this study, we generated a Wrn mutant line that, like humans, relies entirely on dietary sources of vitamin C (ascorbate) to survive, by crossing them to mice that lack the gulonolactone oxidase enzyme required for ascorbate synthesis. In the presence of 0.01% ascorbate (w/v) in drinking water, double-mutant mice exhibited a severe reduction in lifespan, small size, sterility, osteopenia, and metabolic profiles different from wild-type (WT) mice. Although increasing the dose of ascorbate to 0.4% improved dramatically the phenotypes of double-mutant mice, the metabolic and cytokine profiles were different from age-matched WT mice. Finally, double-mutant mice treated with 0.01% ascorbate revealed a permanent activation of all the 3 branches of the ER stress response pathways due to a severe chronic oxidative stress in the ER compartment. In addition, markers associated with the ubiquitin-proteasome-dependent ER-associated degradation pathway were increased. Augmenting the dose of ascorbate reversed the activation of this pathway to WT levels rendering this pathway a potential therapeutic target in WS.-Aumailley, L., Dubois, M. J., Brennan, T. A., Garand, C., Paquet, E. R., Pignolo, R. J., Marette, A., Lebel, M. Serum vitamin C levels modulate the lifespan and endoplasmic reticulum stress response pathways in mice synthesizing a nonfunctional mutant WRN protein.


Subject(s)
Ascorbic Acid/blood , Endoplasmic Reticulum Stress , Longevity , Werner Syndrome Helicase/genetics , Werner Syndrome/metabolism , Animals , Ascorbic Acid/therapeutic use , Female , Loss of Function Mutation , Male , Mice , Mice, Inbred C57BL , Werner Syndrome/drug therapy , Werner Syndrome/genetics
11.
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
12.
Mutagenesis ; 33(4): 301-310, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30137433

ABSTRACT

Reactive oxygen species generate 8-oxo-7,8-dihydroguanine (GO, 8-hydroxyguanine), which induces G:C→T:A transversion mutations. The knockdowns of the protein responsible for Werner syndrome (WRN), a cancer-associated DNA helicase, and DNA polymerase (pol) λ, a WRN-interacting DNA pol, cause untargeted base-substitution mutations (action-at-a-distance mutations). To examine the consequences of the dual reductions of WRN and pol λ for the mutations caused by GO, siRNAs against both proteins were introduced into human U2OS cells. A replicable plasmid DNA with the oxidised nucleobase in a unique position of the supF gene was then introduced into the double knockdown cells. The amplified DNA recovered from the cells was used to transform a bacterial indicator strain. The mutant frequency and the subsequent sequence analysis revealed that the double knockdown additively promoted the G:C→T:A substitution at the GO position and increased the action-at-a-distance mutations to a level similar to that of the single WRN knockdown. Thus, WRN and DNA pol λ seem to suppress the targeted G:C→T:A mutation at least in part independently and reduce the untargeted mutations via an identical pathway.


Subject(s)
DNA Polymerase beta/metabolism , Guanine/analogs & derivatives , Mutation/drug effects , Werner Syndrome/metabolism , DNA/drug effects , DNA Helicases/metabolism , DNA Repair/drug effects , DNA Replication/drug effects , DNA-Directed DNA Polymerase/metabolism , Guanine/pharmacology , Humans , Plasmids/metabolism , Werner Syndrome Helicase/metabolism
13.
Dev Growth Differ ; 58(1): 116-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26691051

ABSTRACT

Progeria is a devastating disorder in which patients exhibit signs of premature aging. The most well-known progeroid syndromes include Hutchinson-Gilford Progeria Syndrome (HGPS) and Werner Syndrome (WS). While HGPS and WS are rare, they often result in severe age-associated complications starting in the early developmental period or after the pubertal growth spurt during adolescence, respectively. In addition, patients with HGPS ultimately die of diseases normally seen in the elderly population, with stroke and myocardial infarction as the leading causes of death. Many WS patients develop similar cardiovascular complications but also have an increased predisposition to developing multiple rare malignancies. These premature aging disorders, as well as animal and cell culture models that recapitulate these diseases, have provided insight into the genetics and cellular pathways that underlie these human conditions and have also uncovered possible mechanisms behind normal aging. Here we discuss the history, the types of progeria, and the various pathophysiological mechanisms that drive these diseases. We also address recent medical advances and treatment modalities for patients with progeria.


Subject(s)
Adolescent Development , Progeria , Puberty , Werner Syndrome , Adolescent , Animals , Female , Humans , Male , Progeria/genetics , Progeria/metabolism , Progeria/physiopathology , Progeria/therapy , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome/physiopathology , Werner Syndrome/therapy
14.
Exp Dermatol ; 25 Suppl 3: 20-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27539898

ABSTRACT

Atypical progeroid syndrome (APS), including atypical Werner syndrome (AWS), is a disorder of premature ageing caused by mutation of the lamin A gene, the same causal gene involved in Hutchinson-Gilford syndrome (HGS). We previously reported the first Japanese case of APS/AWS with a LMNA mutation (p.D300N). Recently, it has been reported that UVA induced abnormal truncated form of lamin A, called progerin, as well as HGS-like abnormal nuclear structures in normal human fibroblasts, being more frequent in the elderly, suggesting that lamin A may be involved in the regulation of photoageing. The objective of this study was to elucidate the sensitivity to cell damage induced by oxidative stress or UVA in fibroblasts from APS/AWS patient. Using immunofluorescence staining and flow cytometry analysis, the amount of early apoptotic cells and degree of intra-cellular reactive oxygen species (ROS) generation were higher in H2 02 - or UVA-treated APS/AWS fibroblasts than in normal fibroblasts, suggesting that repeated UV exposure may induce premature ageing of the skin in APS/AWS patients and that protecting against sunlight is possibly important for delaying the emergence of APS/AWS symptoms. In addition, we demonstrated that H2 O2 -, or UVA-induced apoptosis and necrosis in normal and APS/AWS fibroblasts were enhanced by farnesyltransferase inhibitor (FTI) treatment, indicating that FTI might not be useful for treating our APS/AWS patient.


Subject(s)
Lamin Type A/genetics , Mutation, Missense , Werner Syndrome/genetics , Werner Syndrome/pathology , Amino Acid Substitution , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/radiation effects , Humans , Hydrogen Peroxide/toxicity , Necrosis , Oxidative Stress , Quinolones/pharmacology , Skin/metabolism , Skin/pathology , Skin/radiation effects , Ultraviolet Rays/adverse effects , Werner Syndrome/metabolism
16.
Org Biomol Chem ; 14(3): 947-56, 2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26611938

ABSTRACT

Microwave-assisted synthesis of the pyrazolyl ketone p38 MAPK inhibitor RO3201195 in 7 steps and 15% overall yield, and the comparison of its effect upon the proliferation of Werner Syndrome cells with a library of pyrazolyl ketones, strengthens the evidence that p38 MAPK inhibition plays a critical role in modulating premature cellular senescence in this progeroid syndrome and the reversal of accelerated ageing observed in vitro on treatment with SB203580.


Subject(s)
Ketones/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Small Molecule Libraries/pharmacology , Werner Syndrome/pathology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Ketones/chemical synthesis , Ketones/chemistry , Microwaves , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Werner Syndrome/drug therapy , Werner Syndrome/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Biochim Biophys Acta ; 1842(12 Pt A): 2387-94, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25257404

ABSTRACT

Reduced autophagy may be associated with normal and pathological aging. Here we report a link between autophagy and Werner protein (WRNp), mutated in Werner syndrome, the human premature aging Werner syndrome (WS). WRN mutant fibroblast AG11395 and AG05229 respond weakly to starvation induced autophagy compared to normal cells. While the fusion of phagosomes with lysosome is normal, WS cells contain fewer autophagy vacuoles. Cellular starvation autophagy in WS cells is restored after transfection with full length WRN. Further, siRNA mediated silencing of WRN in the normal fibroblast cell line WI-38 results in decreased autophagy and altered expression of autophagy related proteins. Thus, our observations suggest that WRN may have a role in controlling autophagy and hereby cellular maintenance.


Subject(s)
Autophagy/genetics , Exodeoxyribonucleases/genetics , Gene Expression , RecQ Helicases/genetics , Adult , Autophagy/drug effects , Blotting, Western , Cell Line , Cells, Cultured , Culture Media/chemistry , Culture Media/pharmacology , Exodeoxyribonucleases/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phosphorylation , RNA Interference , RecQ Helicases/metabolism , TOR Serine-Threonine Kinases/metabolism , Transfection , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome/pathology , Werner Syndrome Helicase
18.
Cell Mol Life Sci ; 71(22): 4373-86, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25080110

ABSTRACT

Cellular senescence, a permanent state of cell cycle arrest accompanied by a complex phenotype, is an essential mechanism that limits tumorigenesis and tissue damage. In physiological conditions, senescent cells can be removed by the immune system, facilitating tumor suppression and wound healing. However, as we age, senescent cells accumulate in tissues, either because an aging immune system fails to remove them, the rate of senescent cell formation is elevated, or both. If senescent cells persist in tissues, they have the potential to paradoxically promote pathological conditions. Cellular senescence is associated with an enhanced pro-survival phenotype, which most likely promotes persistence of senescent cells in vivo. This phenotype may have evolved to favor facilitation of a short-term wound healing, followed by the elimination of senescent cells by the immune system. In this review, we provide a perspective on the triggers, mechanisms and physiological as well as pathological consequences of senescent cells.


Subject(s)
Cellular Senescence , Cell Transformation, Neoplastic , DNA Damage , Extracellular Matrix/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Progeria/metabolism , Progeria/physiopathology , Signal Transduction , Werner Syndrome/metabolism , Werner Syndrome/physiopathology
19.
Biochim Biophys Acta ; 1833(12): 3338-3345, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24050918

ABSTRACT

From the surrounding shell to the inner machinery, nuclear proteins provide the functional plasticity of the nucleus. This study highlights the nuclear association of Pore membrane (POM) protein NDC1 and Werner protein (WRN), a RecQ helicase responsible for the DNA instability progeria disorder, Werner Syndrome. In our previous publication, we connected the DNA damage sensor Werner's Helicase Interacting Protein (WHIP), a binding partner of WRN, to the NPC. Here, we confirm the association of the WRN/WHIP complex and NDC1. In established WRN/WHIP knockout cell lines, we further demonstrate the interdependence of WRN/WHIP and Nucleoporins (Nups). These changes do not completely abrogate the barrier of the Nuclear Envelope (NE) but do affect the distribution of FG Nups and the RAN gradient, which are necessary for nuclear transport. Evidence from WRN/WHIP knockout cell lines demonstrates changes in the processing and nucleolar localization of lamin B1. The appearance of "RAN holes" void of RAN corresponds to regions within the nucleolus filled with condensed pools of lamin B1. From WRN/WHIP knockout cell line extracts, we found three forms of lamin B1 that correspond to mature holoprotein and two potential post-translationally modified forms of the protein. Upon treatment with topoisomerase inhibitors lamin B1 cleavage occurs only in WRN/WHIP knockout cells. Our data suggest the link of the NDC1 and WRN as one facet of the network between the nuclear periphery and genome stability. Loss of WRN complex leads to multiple alterations at the NPC and the nucleolus.


Subject(s)
Lamin Type B/metabolism , Nuclear Pore/metabolism , Werner Syndrome/metabolism , Animals , Blotting, Western , Chickens , DNA Damage , DNA Repair , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Gene Knockout Techniques , Membrane Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Binding
20.
BMC Ophthalmol ; 14: 31, 2014 Mar 12.
Article in English | MEDLINE | ID: mdl-24620826

ABSTRACT

BACKGROUND: To present our findings in a case of Werner syndrome with refractory cystoid macular edema (CME) and to determine the expression and the distribution of WRN proteins in human retinas. CASE PRESENTATION: A 35-year-old man with Werner syndrome who developed CME after YAG laser treatment was studied. Optical coherence tomographic (OCT) scans were used to examine the CME in the right eye. The patient received topical eye drops (0.1% bromfenac sodium hydrate twice daily and 1% dorzolamide hydrochloride thrice daily), sub-Tenon triamcinolone injection thrice, intravitreal bevacizumab injection twice, and pars plana vitrectomy of the right eye. Genetic analyses were performed to diagnose the disease. To examine the expression and distribution of WRN proteins in the retinas, immunohistochemistry for WRN proteins was performed in human retinas. The CME in the right eye was not improved by any of the treatments. During the follow-up period, CME developed in the left eye. Genetic analyses detected compound heterozygosity, Mut4 and Mut11, in the WRN gene and the individual was diagnosed with Werner syndrome. Immunohistochemical analysis of WRN proteins expression in human retinas showed that WRN proteins were expressed in the parts of the Müller cells in the inner nuclear layer and outer nuclear layer. CONCLUSION: Patients with Werner syndrome can develop severe CME after laser treatment. A pathological link may exist between mutations in the WRN gene and the development of CME in patients with Werner syndrome.


Subject(s)
Exodeoxyribonucleases/metabolism , Macular Edema/metabolism , RecQ Helicases/metabolism , Retina/metabolism , Werner Syndrome/metabolism , Adult , Humans , Immunohistochemistry , Laser Therapy/adverse effects , Male , Werner Syndrome Helicase
SELECTION OF CITATIONS
SEARCH DETAIL