Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
1.
Antioxid Redox Signal ; 37(1-3): 208-228, 2022 07.
Article in English | MEDLINE | ID: mdl-34428933

ABSTRACT

Significance: Reactive species have been classically considered causative of age-related degenerative processes, but the scenario appears considerably more complex and to some extent counterintuitive than originally anticipated. The impact of reactive species in precocious aging syndromes is revealing new clues to understand and perhaps challenge the resulting degenerative processes. Recent Advances: Our understanding of reactive species has considerably evolved, including their hormetic effect (beneficial at a certain level, harmful beyond this level), the occurrence of diverse hormetic peaks in different cell types and organisms, and the extended type of reactive species that are relevant in biological processes. Our understanding of the impact of reactive species has also expanded from the dichotomic damaging/signaling role to modulation of gene expression. Critical Issues: These new concepts are affecting the study of aging and diseases where aging is greatly accelerated. We discuss how notions arising from the study of the underlying mechanisms of a progeroid disease, Cockayne syndrome, represent a paradigm shift that may shed a new light in understanding the role of reactive species in age-related degenerative processes. Future Issues: Future investigations urge to explore established and emerging notions to elucidate the multiple contributions of reactive species in degenerative processes linked to pathophysiological aging and their possible amelioration. Antioxid. Redox Signal. 37, 208-228.


Subject(s)
Aging , Cockayne Syndrome , Reactive Nitrogen Species , Reactive Oxygen Species , Sulfur , Animals , Antioxidants/therapeutic use , Cockayne Syndrome/physiopathology , Down Syndrome/physiopathology , Humans , Mitochondria , Oxidative Stress , Progeria/physiopathology , Reactive Nitrogen Species/chemistry , Reactive Oxygen Species/chemistry , Signal Transduction , Sulfur/chemistry , Werner Syndrome/physiopathology
2.
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
4.
Aging (Albany NY) ; 12(24): 24940-24956, 2020 12 29.
Article in English | MEDLINE | ID: mdl-33373317

ABSTRACT

Patients with Werner syndrome present with diverse signs of aging that begin in adolescence. A Japanese nationwide survey was conducted to establish a registry that could clarify the disease profile of patients with Werner syndrome. The questionnaires were sent to 7888 doctors. The survey identified 116 patients diagnosed with Werner syndrome based on the diagnosis criteria. Forty patients were enrolled in the registry. Data on clinical symptoms, treatment information, and laboratory examination from patients who provided informed consent were collected. The data at enrollment were analyzed. The patients' average age at enrollment was 50.1±7.5 years. The mean onset age was 26.1±9.5 years, but the mean age at diagnosis was 42.5±8.6 years. Average height and weight of the study patients were lower than those of Japanese individuals. Almost all patients experienced hair change and cataracts. More than 60% of patients presented with glycolipid abnormalities. Overall, 15% of patients had a history of foot amputation. Approximately 30% of the patients' parents had a consanguineous marriage. The average grip strength, walking speed, and skeletal muscle mass index met the diagnostic criteria for sarcopenia. The registry revealed that there are opportunities for early diagnosis and intervention; therefore, sensitization about the disease is needed.


Subject(s)
Delayed Diagnosis/statistics & numerical data , Werner Syndrome/diagnosis , Adolescent , Adult , Age of Onset , Alopecia/physiopathology , Calcinosis/physiopathology , Cataract/physiopathology , Consanguinity , Diabetes Mellitus , Dyslipidemias , Early Diagnosis , Early Medical Intervention , Fatty Liver , Female , Hair Color , Hand Strength , Humans , Japan , Male , Middle Aged , Pigmentation Disorders/physiopathology , Sarcopenia/physiopathology , Skin Ulcer/physiopathology , Walking Speed , Werner Syndrome/physiopathology , Young Adult
5.
Exp Gerontol ; 127: 110733, 2019 11.
Article in English | MEDLINE | ID: mdl-31518666

ABSTRACT

Werner syndrome (WS) is an autosomal recessive progeroid disease characterized by patients' early onset of aging, increased risk of cancer and other age-related pathologies. WS is caused by mutations in WRN, a RecQ helicase that has essential roles responding to DNA damage and preventing genomic instability. While human WRN has both an exonuclease and helicase domain, Drosophila WRNexo has high genetic and functional homology to only the exonuclease domain of WRN. Like WRN-deficient human cells, Drosophila WRNexo null mutants (WRNexoΔ) are sensitive to replication stress, demonstrating mechanistic similarities between these two models. Compared to age-matched wild-type controls, WRNexoΔ flies exhibit increased physiological signs of aging, such as shorter lifespans, higher tumor incidence, muscle degeneration, reduced climbing ability, altered behavior, and reduced locomotor activity. Interestingly, these effects are more pronounced in females suggesting sex-specific differences in the role of WRNexo in aging. This and future mechanistic studies will contribute to our knowledge in linking faulty DNA repair mechanisms with the process of aging.


Subject(s)
Aging, Premature/genetics , Drosophila Proteins/deficiency , Exonucleases/deficiency , Werner Syndrome/physiopathology , Aging, Premature/physiopathology , Animals , Behavior, Animal/physiology , Body Composition/physiology , Body Weight/physiology , DNA Repair/physiology , Drosophila , Drosophila Proteins/genetics , Exonucleases/genetics , Female , Gastrointestinal Neoplasms/physiopathology , Male , Motor Activity/physiology , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Mutation/genetics , Phenotype
6.
Endocr J ; 66(11): 961-969, 2019 Nov 28.
Article in English | MEDLINE | ID: mdl-31270292

ABSTRACT

Werner syndrome (WS) is a rare, adult-onset progeroid syndrome. Classic WS is caused by WRN mutation and partial atypical WS (AWS) is caused by LMNA mutation. A 19-year-old female patient with irregular menstruation and hyperglycemia was admitted. Physical examination revealed characteristic faces of progeria, graying and thinning of the hair scalp, thinner and atrophic skin over the hands and feet, as well as lipoatrophy of the extremities, undeveloped breasts at Tanner stage 3, and short stature. The patient also suffered from severe insulin-resistant diabetes mellitus, hyperlipidemia, fatty liver, and polycystic ovarian morphology. Possible WS was considered and both WRN and LMNA genes were analyzed. A novel missense mutation p.L140Q (c.419T>A) in the LMNA gene was identified and confirmed the diagnosis of AWS. Her father was a carrier of the same mutation. We carried out therapy for lowering blood glucose and lipid and improving insulin resistance, et al. The fasting glucose, postprandial glucose and triglyceride level was improved after treatment for 9 days. Literature review of AWS was performed to identify characteristics of the disease. Diabetes mellitus is one of the clinical manifestations of WS and attention must give to the differential diagnosis. Gene analysis is critical in the diagnosis of WS. According to the literature, classic and atypical WS differ in incidence, pathogenic gene, and clinical manifestations. Characteristic dermatological pathology may be significantly more important for the initial identification of AWS. Early detection, appropriate treatments, and regular follow-up may improve prognosis and survival of WS patients.


Subject(s)
Diabetes Mellitus/metabolism , Hypogonadism/physiopathology , Werner Syndrome/physiopathology , Diabetes Mellitus/etiology , Diabetes Mellitus/genetics , Female , Humans , Hypogonadism/etiology , Hypogonadism/genetics , Hypogonadism/metabolism , Lamin Type A/genetics , Werner Syndrome/complications , Werner Syndrome/genetics , Young Adult
7.
Brain Behav Immun ; 73: 450-469, 2018 10.
Article in English | MEDLINE | ID: mdl-29908963

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 lifespan. Yet, little is known about the impact of WRN mutations on the central nervous system in both humans and mouse models of WS. In the current study, we have performed a longitudinal behavioral assessment on mice bearing a Wrn helicase deletion. Behavioral tests demonstrated a loss of motor activity and coordination, reduction in perception, increase in repetitive behavior, and deficits in both spatial and social novelty memories in Wrn mutant mice compared to age-matched wild type mice. These neurological deficits were associated with biochemical and histological changes in the brain of aged Wrn mutant mice. Microglia, resident immune cells that regulate neuronal plasticity and function in the brain, were hyper-ramified in multiple regions involved with the behavioral deficits of Wrn mutant mice. Furthermore, western analyses indicated that Wrn mutant mice exhibited an increase of oxidative stress markers in the prefrontal cortex. Supporting these findings, electron microscopy studies revealed increased cellular aging and oxidative stress features, among microglia and neurons respectively, in the prefrontal cortex of aged Wrn mutant mice. In addition, multiplex immunoassay of serum identified significant changes in the expression levels of several pro- and anti-inflammatory cytokines. Taken together, these findings indicate that microglial dysfunction and neuronal oxidative stress, associated with peripheral immune system alterations, might be important driving forces leading to abnormal neurological symptoms in WS thus suggesting potential therapeutic targets for interventions.


Subject(s)
Werner Syndrome Helicase/physiology , Werner Syndrome/genetics , Animals , Cellular Senescence/physiology , DNA Damage/physiology , Disease Models, Animal , Female , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Motor Activity/genetics , Motor Activity/physiology , Mutant Proteins , Neurons/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Werner Syndrome/immunology , Werner Syndrome/physiopathology , Werner Syndrome Helicase/genetics
8.
DNA Repair (Amst) ; 68: 1-11, 2018 08.
Article in English | MEDLINE | ID: mdl-29800817

ABSTRACT

Impaired autophagy may be associated with normal and pathological aging. Here we explore a link between autophagy and domain function of Werner protein (WRNp). Werner (WRN) mutant cell lines AG11395, AG05229 and normal aged fibroblast AG13129 display a deficient response to tunicamycin mediated endoplasmic reticulum (ER) stress induced autophagy compared to clinically unaffected GM00637 and normal young fibroblast GM03440. Cellular endoplasmic reticulum (ER) stress mediated autophagy in WS and normal aged cells is restored after transfection with wild type full length WRN, but deletion of the acidic domain from wild type WRN fails to restore autophagy. The acidic domain of WRNp was shown to regulate its transcriptional activity, and here, we show that it affects the transcription of certain proteins involved in autophagy and aging. Furthermore, siRNA mediated silencing of WRN in normal fibroblast WI-38 resulted in decrease of age related proteins Lamin A/C and Mre11.


Subject(s)
Autophagy , Endoplasmic Reticulum Stress , Protein Domains , Werner Syndrome Helicase/metabolism , Werner Syndrome/metabolism , Adolescent , Adult , Aged, 80 and over , Cell Line , Female , Gene Expression Regulation , Humans , Lamins/genetics , MRE11 Homologue Protein/genetics , Male , Middle Aged , Mutation , Up-Regulation , Werner Syndrome/physiopathology , Werner Syndrome Helicase/genetics , Werner Syndrome Helicase/physiology , Young Adult
9.
Protein Cell ; 9(4): 333-350, 2018 04.
Article in English | MEDLINE | ID: mdl-29476423

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) and Werner syndrome (WS) are two of the best characterized human progeroid syndromes. HGPS is caused by a point mutation in lamin A (LMNA) gene, resulting in the production of a truncated protein product-progerin. WS is caused by mutations in WRN gene, encoding a loss-of-function RecQ DNA helicase. Here, by gene editing we created isogenic human embryonic stem cells (ESCs) with heterozygous (G608G/+) or homozygous (G608G/G608G) LMNA mutation and biallelic WRN knockout, for modeling HGPS and WS pathogenesis, respectively. While ESCs and endothelial cells (ECs) did not present any features of premature senescence, HGPS- and WS-mesenchymal stem cells (MSCs) showed aging-associated phenotypes with different kinetics. WS-MSCs had early-onset mild premature aging phenotypes while HGPS-MSCs exhibited late-onset acute premature aging characterisitcs. Taken together, our study compares and contrasts the distinct pathologies underpinning the two premature aging disorders, and provides reliable stem-cell based models to identify new therapeutic strategies for pathological and physiological aging.


Subject(s)
Aging/genetics , Human Embryonic Stem Cells/metabolism , Progeria/genetics , Werner Syndrome/genetics , Aging/physiology , DNA Helicases/genetics , Human Embryonic Stem Cells/physiology , Humans , Kinetics , Lamin Type A/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mutation , Progeria/physiopathology , Werner Syndrome/physiopathology
10.
Curr Aging Sci ; 11(1): 55-62, 2018.
Article in English | MEDLINE | ID: mdl-29149836

ABSTRACT

BACKGROUND: Most syndromes of accelerated aging are caused by mutations affecting the integrity of the genetic material. Among them, the most studied is Werner's syndrome, "adult progeria", caused by a recessive autosomal mutation with a frequency of 1 in 10 million, which affects a helicase involved in DNA repair. In Werner syndrome, there is a loss of heterochromatin, though the stability of heterochromatin is also affected in "normal" aging. The Hutchinson-Gilford Progeria Syndrome (HGPS), "child progeria", has an even lower frequency. In most cases, it is caused by a point mutation of a gene coding a protein in the nuclear envelope, lamin A. OBJECTIVES: HGPS may provide valuable insights into the aging process. The symptoms of this condition do not entirely overlap with those of "normal" aging. METHOD: A critical analysis of the accelerated aging syndromes may explain what aging is, and also why some tissues and organs age at accelerated rates as compared to other tissues. RESULTS: In this article, we will discuss the implications of HGPS and other accelerated aging syndromes in the light of the biochemical hypothesis of aging we advanced. According to this hypothesis, some reactions are less stimulated and diminish in time, affecting not only specific biochemical functions, but cellular energy, and therefore its capacity for synthesis. CONCLUSION: Besides, a new vision on aging, possible therapeutic strategies for these conditions and others, with similar mechanisms, are also presented.


Subject(s)
Aging/genetics , Lamin Type A/genetics , Mutation , Progeria/genetics , Werner Syndrome Helicase/genetics , Werner Syndrome/genetics , Age Factors , Aging/pathology , Animals , Chromatin Assembly and Disassembly , Energy Metabolism/genetics , Fibrosis , Genetic Predisposition to Disease , Humans , Models, Genetic , Phenotype , Progeria/pathology , Progeria/physiopathology , Werner Syndrome/pathology , Werner Syndrome/physiopathology
11.
Ageing Res Rev ; 33: 105-114, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26993153

ABSTRACT

Werner syndrome (WS) is a prototypical segmental progeroid syndrome characterized by multiple features consistent with accelerated aging. It is caused by null mutations of the WRN gene, which encodes a member of the RECQ family of DNA helicases. A unique feature of the WRN helicase is the presence of an exonuclease domain in its N-terminal region. Biochemical and cell biological studies during the past decade have demonstrated involvements of the WRN protein in multiple DNA transactions, including DNA repair, recombination, replication and transcription. A role of the WRN protein in telomere maintenance could explain many of the WS phenotypes. Recent discoveries of new progeroid loci found in atypical Werner cases continue to support the concept of genomic instability as a major mechanism of biological aging. Based on these biological insights, efforts are underway to develop therapeutic interventions for WS and related progeroid syndromes.


Subject(s)
Aging, Premature , Werner Syndrome Helicase/genetics , Werner Syndrome , Aging, Premature/genetics , Aging, Premature/metabolism , DNA Repair , DNA Replication , Exodeoxyribonucleases , Humans , Mutation , Werner Syndrome/diagnosis , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome/physiopathology
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.
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
16.
Bull Math Biol ; 76(6): 1241-69, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24771273

ABSTRACT

Human cells typically consist of 23 pairs of chromosomes. Telomeres are repetitive sequences of DNA located at the ends of chromosomes. During cell replication, a number of basepairs are lost from the end of the chromosome and this shortening restricts the number of divisions that a cell can complete before it becomes senescent, or non-replicative. In this paper, we use Monte Carlo simulations to form a stochastic model of telomere shortening to investigate how telomere shortening affects normal aging. Using this model, we study various hypotheses for the way in which shortening occurs by comparing their impact on aging at the chromosome and cell levels. We consider different types of length-dependent loss and replication probabilities to describe these processes. After analyzing a simple model for a population of independent chromosomes, we simulate a population of cells in which each cell has 46 chromosomes and the shortest telomere governs the replicative potential of the cell. We generalize these simulations to Werner's syndrome, a condition in which large sections of DNA are removed during cell division and, amongst other conditions, results in rapid aging. Since the mechanisms governing the loss of additional basepairs are not known, we use our model to simulate a variety of possible forms for the rate at which additional telomeres are lost per replication and several expressions for how the probability of cell division depends on telomere length. As well as the evolution of the mean telomere length, we consider the standard deviation and the shape of the distribution. We compare our results with a variety of data from the literature, covering both experimental data and previous models. We find good agreement for the evolution of telomere length when plotted against population doubling.


Subject(s)
Aging/physiology , Chromosomes/physiology , Models, Biological , Telomere/physiology , Werner Syndrome/physiopathology , Cell Division , Computer Simulation , DNA Replication , Humans , Monte Carlo Method , Stochastic Processes
17.
Exp Gerontol ; 47(12): 936-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960593

ABSTRACT

Minor inflammation-driven aging (inflammaging) has been proposed to explain human aging mechanism. To study the inflammatory condition associated with normal human aging, highly sensitive CRP (hsCRP) was examined in the sera collected from 217 healthy Japanese individuals aged between 1 and 100years and 41 mutation-proven Japanese Werner syndrome (WS) patients. The serum hsCRP was assayed by ELISA. The serum hsCRP level increased significantly (p<0.001) with normal aging from both sexes. The serum hsCRP was significantly elevated in WS (mean±SE: 11.0±1.6µg/ml) compared with age-matched normal population (1.3±0.3µg/ml, p<0.001) and normal elderly population ages between 71 and 100years (4.2±0.7µg/ml, p<0.001). Both normal aging and WS were associated with minor inflammation that can be evaluated by serum hsCRP. WS may be a good candidate to study inflammaging.


Subject(s)
Aging/physiology , Inflammation/physiopathology , Werner Syndrome/physiopathology , Adolescent , Adult , Aged , Aging/blood , Biomarkers/blood , C-Reactive Protein/metabolism , Child , Child, Preschool , Female , Humans , Infant , Inflammation/blood , Inflammation/complications , Male , Middle Aged , Werner Syndrome/blood , Werner Syndrome/complications , Young Adult
18.
Geriatr Gerontol Int ; 12(1): 140-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22188495

ABSTRACT

Werner syndrome (WS) is an autosomal recessive progeroid disorder caused by mutations in the WRN DNA helicase. It is characterized by the graying and loss of hair, juvenile cataracts, sclerosis and ulceration of skin, insulin-resistant diabetes mellitus, dyslipidemia, abdominal adiposity, osteoporosis, atherosclerosis, and malignant neoplasm. Patients are usually diagnosed in their 30s or 40s, but the early pathophysiology of the syndrome is still not fully understood. Here we report a 29-year-old female patient who displayed cataracts, hair graying, and tendinous calcinosis. Her parents were first cousins. Interestingly, the patient lacked the metabolic signs typical for WS, including glucose intolerance, dyslipidemia, and visceral fat accumulation. A hyperinsulinemic response at 30 min was observed in an oral glucose tolerance test. Mutational analysis for the WRN gene revealed a homozygous nucleotide substitution 3190C>T in exon 24, resulting in a protein product with replacement of an arginine residue at position 573 by termination codon (Arg987Ter). The mutated WRN protein was unable to translocate into the nucleus in an in vitro cell assay. A WS patient with an Arg987Ter mutation has been previously reported in Switzerland, the present case is the first to be identified in Asia. This case demonstrates the early clinical features of WS and suggests that metabolic abnormality, including insulin resistance, is not an essential component of WS at disease onset. Moreover, a follow-up study of such case would be useful to understand how the various clinical symptoms in WS develop and progress over the years.


Subject(s)
Insulin Resistance/physiology , Werner Syndrome/physiopathology , Adult , DNA Mutational Analysis , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Female , Follow-Up Studies , Humans , Mutation , RecQ Helicases/genetics , RecQ Helicases/metabolism , Werner Syndrome/genetics , Werner Syndrome/metabolism , Werner Syndrome Helicase
19.
Aging (Albany NY) ; 3(9): 889-95, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21931180

ABSTRACT

Hutchinson-Gilford Progeria (HGPS) and Werner syndromes are diseases that clinically resemble some aspects of accelerated aging. HGPS is caused by mutations in theLMNA gene resulting in post-translational processing defects that trigger Progeria in children. Werner syndrome, arising from mutations in the WRN helicase gene, causes premature aging in young adults. What are the molecular mechanism(s) underlying these disorders and what aspects of the diseases resemble physiological human aging? Much of what we know stems from the study of patient derived fibroblasts with both mutations resulting in increased DNA damage, primarily at telomeres. However, in vivo patients with Werner's develop arteriosclerosis, among other pathologies. In HGPS patients, including iPS derived cells from HGPS patients, as well as some mouse models for Progeria, vascular smooth muscle (VSM) appears to be among the most severely affected tissues. Defective Lamin processing, associated with DNA damage, is present in VSM from old individuals, indicating processing defects may be a factor in normal aging. Whether persistent DNA damage, particularly at telomeres, is the root cause for these pathologies remains to be established, since not all progeroid Lmna mutations result in DNA damage and genome instability.


Subject(s)
Aging/physiology , Progeria/physiopathology , Werner Syndrome/physiopathology , Animals , Child , DNA Damage , Disease Models, Animal , Exodeoxyribonucleases/genetics , Genomic Instability , Humans , Lamin Type A/genetics , Mice , Mutation , Progeria/genetics , RecQ Helicases/genetics , Werner Syndrome/genetics , Werner Syndrome Helicase , Young Adult
20.
Aging (Albany NY) ; 3(3): 277-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21464516

ABSTRACT

Werner syndrome (WS) is a rare disorder characterized by the premature onset of several pathologies associated with aging. The gene responsible for WS codes for a RecQ-type DNA helicase and is believed to be involved in different aspects of DNA repair, replication, and transcription. We recently identified the Scaffold attachment factor B1 (SAFB1) as a potential interactants in human cells. SAFB1 is a multifunctional protein that binds both nucleic acids and is involved in the attachment of chromatin to the nuclear matrix, transcription, and stress response. Mice lacking SAFB1 exhibit developmental abnormalities in their lungs, high incidence of perinatal lethality, and adults develop different types of tumors. Mouse embryonic fibroblasts from Safb1-null animals are immortalized in culture. In this study, mice with a mutation in the helicase domain of the Wrn gene were crossed to Safb1-null mice. Double homozygous mutant mice exhibited increased apoptosis, a lower cell proliferation rate in their lungs and a higher incidence of perinatal death compared to Safb1-null mice. Few double homozygous mutants survived weaning and died before the age of six months. Finally, mouse embryonic fibroblasts lacking a functional Wrn helicase inhibited the immortalization of Safb1-null cells. These results indicate that an intact Wrn protein is required for immortalization and tumorigenesis in Safb1-null mice.


Subject(s)
DNA-Binding Proteins/deficiency , RecQ Helicases/physiology , Animals , Apoptosis , Cell Proliferation , Cell Survival , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Exodeoxyribonucleases/physiology , Female , HEK293 Cells , Homozygote , Humans , Longevity , Lung/pathology , Male , Matrix Attachment Region Binding Proteins/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/etiology , Nuclear Matrix-Associated Proteins/physiology , Pregnancy , Protein Kinase C-delta/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/physiology , RecQ Helicases/deficiency , RecQ Helicases/genetics , Receptors, Estrogen/physiology , Werner Syndrome/genetics , Werner Syndrome/pathology , Werner Syndrome/physiopathology , Werner Syndrome Helicase , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...