Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 1296, 2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-31992827

RESUMEN

Mitochondrial mutations and dysfunction have been demonstrated in several age-related disorders including osteoarthritis, yet its relative contribution to pathogenesis remains unknown. Here we evaluated whether premature aging caused by accumulation of mitochondrial DNA mutations in PolgD275A mice predisposes to the development of knee osteoarthritis. Compared with wild type animals, homozygous PolgD275A mice displayed a specific bone phenotype characterized by osteopenia of epiphyseal trabecular bone and subchondral cortical plate. Trabecular thickness was significantly associated with osteocyte apoptosis rates and osteoclasts numbers were increased in subchondral bone tissues. While chondrocyte apoptosis rates in articular and growth plate cartilage were similar between groups, homozygous mitochondrial DNA mutator mice displayed elevated numbers of hypertrophic chondrocytes in articular calcified cartilage. Low grade cartilage degeneration, predominantly loss of proteoglycans, was present in all genotypes and the development of osteoarthritis features was not found accelerated in premature aging. Somatically acquired mitochondrial DNA mutations predispose to elevated subchondral bone turnover and hypertrophy in calcified cartilage, yet additional mechanical or metabolic stimuli would seem required for induction and accelerated progression of aging-associated osteoarthritis.


Asunto(s)
Envejecimiento Prematuro , Enfermedades Óseas Metabólicas , Condrocitos , ADN Polimerasa gamma , Mutación Missense , Osteoartritis , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Sustitución de Aminoácidos , Animales , Enfermedades Óseas Metabólicas/enzimología , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Condrocitos/enzimología , Condrocitos/patología , ADN Polimerasa gamma/genética , ADN Polimerasa gamma/metabolismo , Hipertrofia , Ratones , Ratones Mutantes , Osteoartritis/enzimología , Osteoartritis/genética , Osteoartritis/patología
2.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(4): 415-420, 2018 Apr 28.
Artículo en Chino | MEDLINE | ID: mdl-29774879

RESUMEN

OBJECTIVE: To explore the role ofDNA methyltransferase 1 (DNMT1) in mouse skin aging.
 Methods: Epidermal conditional K14 Cre-mediated DNA methyltransferase 1 (DNMT1) knockout mice (Mut group, n=4) and the littermate normal mice with the same age (WT group) n=4) were used in this study. HE staining was used to detect the pathological changes of skin; the changes of number in the dermal elastic fibers were detected by Gomori aldehyde fuchsin staining, the number of 5-bromo-2-deoxyuridine (BrdU)-labeled transit amplifying cells (TAC) in epidermis were detected by immunohistochemical staining; the number of chlorodeoxyuridine (CldU)-label-retaining cells (LRC) in epidermis were detected by immunofluorescent staining.
 Results: Compared with the WT group, the skin showed premature aging symptoms in the Mut group concomitant with the decreased epidermal thickness as well as the number of dermal collagen fibers, while the increased dermal elastic fiber fracture. Compared with the WT group, the number of TAC in the epidermis was significantly increased (P<0.05), and the number of LRC was significantly decreased (P<0.05) in the Mut group.
 Conclusion: The phenotype of skin premature aging in epidermal stem cell conditional DNMT1-knockout mice suggests an important role of DNMT1 in skin aging.


Asunto(s)
Envejecimiento Prematuro/patología , ADN (Citosina-5-)-Metiltransferasa 1/fisiología , Envejecimiento de la Piel/patología , Envejecimiento Prematuro/enzimología , Animales , ADN (Citosina-5-)-Metiltransferasa 1/genética , Células Epidérmicas , Ratones , Ratones Noqueados , Envejecimiento de la Piel/fisiología
3.
J Biol Chem ; 291(16): 8374-86, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26887940

RESUMEN

Normal human stem cells rely on low levels of active telomerase to sustain their high replicative requirements. Deficiency in telomere maintenance mechanisms leads to the development of premature aging diseases, such as dyskeratosis congenita and aplastic anemia. Mutations in the unique "insertion in fingers domain" (IFD) in the human telomerase reverse transcriptase catalytic subunit (hTERT) have previously been identified and shown to be associated with dyskeratosis congenita and aplastic anemia. However, little is known about the molecular mechanisms impacted by these IFD mutations. We performed comparative functional analyses of disease-associated IFD variants at the molecular and cellular levels. We report that hTERT-P721R- and hTERT-R811C-expressing cells exhibited growth defects likely due to impaired TPP1-mediated recruitment of these variant enzymes to telomeres. We showed that activity and processivity of hTERT-T726M failed to be stimulated by TPP1-POT1 overexpression and that dGTP usage by this variant was less efficient compared with the wild-type enzyme. hTERT-P785L-expressing cells did not show growth defects, and this variant likely confers cell survival through increased DNA synthesis and robust activity stimulation by TPP1-POT1. Altogether, our data suggest that multiple mechanisms contribute to cell growth defects conferred by the IFD variants.


Asunto(s)
Envejecimiento Prematuro/enzimología , Mutación Missense , Telomerasa/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Sustitución de Aminoácidos , Aminopeptidasas/genética , Aminopeptidasas/metabolismo , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Estructura Terciaria de Proteína , Serina Proteasas/genética , Serina Proteasas/metabolismo , Complejo Shelterina , Telomerasa/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
4.
Trends Mol Med ; 21(6): 364-72, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25964054

RESUMEN

The multidomain protein kinase BubR1 is a central component of the mitotic spindle assembly checkpoint (SAC), an essential self-monitoring system of the eukaryotic cell cycle that ensures the high fidelity of chromosome segregation by delaying the onset of anaphase until all chromosomes are properly bi-oriented on the mitotic spindle. We discuss the roles of BubR1 in the SAC and the implications of BubR1-mediated interactions that protect against aneuploidy. We also describe the emerging roles of BubR1 in cellular processes that extend beyond the SAC, discuss how mice models have revealed unanticipated functions for BubR1 in the regulation of normal aging, and the potential role of BubR1 as therapeutic target for the development of innovative anticancer therapies.


Asunto(s)
Envejecimiento Prematuro/enzimología , Aneuploidia , Proteínas Serina-Treonina Quinasas/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Animales , Inestabilidad Genómica , Humanos , Puntos de Control de la Fase M del Ciclo Celular , Modelos Moleculares , Terapia Molecular Dirigida , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética
5.
Protein Cell ; 6(5): 322-33, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25907989

RESUMEN

The sirtuin proteins constitute class III histone deacetylases (HDACs). These evolutionarily conserved NAD(+)-dependent enzymes form an important component in a variety of cellular and biological processes with highly divergent as well as convergent roles in maintaining metabolic homeostasis, safeguarding genomic integrity, regulating cancer metabolism and also inflammatory responses. Amongst the seven known mammalian sirtuin proteins, SIRT1 has gained much attention due to its widely acknowledged roles in promoting longevity and ameliorating age-associated pathologies. The contributions of other sirtuins in the field of aging are also gradually emerging. Here, we summarize some of the recent discoveries in sirtuins biology which clearly implicate the functions of sirtuin proteins in the regulation of premature cellular senescence and accelerated aging. The roles of sirtuins in various cellular processes have been extrapolated to draw inter-linkage with anti-aging mechanisms. Also, the latest findings on sirtuins which might have potential effects in the process of aging have been reviewed.


Asunto(s)
Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Longevidad/genética , Sirtuina 1/genética , Sirtuina 1/metabolismo , Animales , Humanos
6.
Mech Ageing Dev ; 146-148: 42-52, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25843235

RESUMEN

Though defective genome maintenance and DNA repair have long been known to promote phenotypes of premature aging, the role protein methylation plays in these processes is only now emerging. We have recently identified the first N-terminal methyltransferase, NRMT1, which regulates protein-DNA interactions and is necessary for both accurate mitotic division and nucleotide excision repair. To demonstrate if complete loss of NRMT1 subsequently resulted in developmental or aging phenotypes, we constructed the first NRMT1 knockout (Nrmt1(-/-)) mouse. The majority of these mice die shortly after birth. However, the ones that survive, exhibit decreased body size, female-specific infertility, kyphosis, decreased mitochondrial function, and early-onset liver degeneration; phenotypes characteristic of other mouse models deficient in DNA repair. The livers from Nrmt1(-/-) mice produce less reactive oxygen species (ROS) than wild type controls, and Nrmt1(-/-) mouse embryonic fibroblasts show a decreased capacity for handling oxidative damage. This indicates that decreased mitochondrial function may benefit Nrmt1(-/-) mice and protect them from excess internal ROS and subsequent DNA damage. These studies position the NRMT1 knockout mouse as a useful new system for studying the effects of genomic instability and defective DNA damage repair on organismal and tissue-specific aging.


Asunto(s)
Envejecimiento Prematuro , Reparación del ADN , Metiltransferasas/deficiencia , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Animales , Femenino , Fibroblastos/enzimología , Fibroblastos/patología , Infertilidad Femenina/enzimología , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo
7.
Brain Res ; 1552: 41-54, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24457043

RESUMEN

ß-Asarone is an active component of the Acori graminei rhizome that is a traditional Chinese medicine clinically used in treating dementia in China. However, the cognitive effect of ß-asarone and its mechanism has remained elusive. Here, we used asenescence-accelerated prone 8 (SAMP8) mice, which mimic many of the salient features of Alzheimer׳s disease (AD), to further investigate whether modulation of the ROCK signaling pathway and/or autophagy, synaptic loss is involved in the effects of ß-asarone on learning and memory. SAMP8 mice at the age of 6 months were intragastrically administered by ß-asarone or a vehicle daily for 2 months. Senescence-accelerated-resistant (SAMR1) mice were used as the control. Our results demonstrate that autophagy and ROCK expression were increased significantly in 8 months SAMP8 mice, which were concomitant with that SAMP8 mice at the same age displayed a significant synaptic loss and cognitive deficits. The up-regulation of ROCK expression and autophage in the hippocampus of SAMP8 were significantly reduced by ß-asarone, and prevents synaptic loss and improved cognitive function of the SAMP8 mice. ß-asarone decreased neuronophagia and lipofuscin in the hippocampus of SAMP8 mice, but did not reduce Aß42 levels and malondialdehyde levels and superoxide dismutase activities. Moreover, suppression of ROCK2 by siRNA significantly reduced the effects of ß-asarone on the autophage and synaptic proteins expression in PC12 cells damage induced by Aß1-40. Taken together, ß-asarone prevents autophagy and synaptic loss by reducing ROCK expression in SAMP8 mice.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Anisoles/uso terapéutico , Autofagia/efectos de los fármacos , Región CA3 Hipocampal/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Sinapsis/efectos de los fármacos , Quinasas Asociadas a rho/biosíntesis , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/psicología , Derivados de Alilbenceno , Péptidos beta-Amiloides/análisis , Animales , Anisoles/farmacología , Región CA3 Hipocampal/química , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/prevención & control , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Lipofuscina/análisis , Potenciación a Largo Plazo/efectos de los fármacos , Malondialdehído/análisis , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/análisis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Células PC12 , Fragmentos de Péptidos/análisis , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Ratas , Superóxido Dismutasa/análisis , Sinapsis/enzimología , Regulación hacia Arriba/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/fisiología
8.
Aging Cell ; 12(5): 802-13, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23738891

RESUMEN

The ubiquitin-proteasome system is central to the regulation of cellular proteostasis. Nevertheless, the impact of in vivo proteasome dysfunction on the proteostasis networks and the aging processes remains poorly understood. We found that RNAi-mediated knockdown of 20S proteasome subunits in Drosophila melanogaster resulted in larval lethality. We therefore studied the molecular effects of proteasome dysfunction in adult flies by developing a model of dose-dependent pharmacological proteasome inhibition. Impaired proteasome function promoted several 'old-age' phenotypes and markedly reduced flies' lifespan. In young somatic tissues and in gonads of all ages, loss of proteasome activity induced higher expression levels and assembly rates of proteasome subunits. Proteasome dysfunction was signaled to the proteostasis network by reactive oxygen species that originated from malfunctioning mitochondria and triggered an Nrf2-dependent upregulation of the proteasome subunits. RNAi-mediated Nrf2 knockdown reduced proteasome activities, flies' resistance to stress, as well as longevity. Conversely, inducible activation of Nrf2 in transgenic flies upregulated basal proteasome expression and activity independently of age and conferred resistance to proteotoxic stress. Interestingly, prolonged Nrf2 overexpression reduced longevity, indicating that excessive activation of the proteostasis pathways can be detrimental. Our in vivo studies add new knowledge on the proteotoxic stress-related regulation of the proteostasis networks in higher metazoans. Proteasome dysfunction triggers the activation of an Nrf2-dependent tissue- and age-specific regulatory circuit aiming to adjust the cellular proteasome activity according to temporal and/or spatial proteolytic demands. Prolonged deregulation of this proteostasis circuit accelerates aging.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Deficiencias en la Proteostasis/metabolismo , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/prevención & control , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Drosophila melanogaster/enzimología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Masculino , Factor 2 Relacionado con NF-E2/genética , Estrés Oxidativo/fisiología , Deficiencias en la Proteostasis/genética , Especies Reactivas de Oxígeno/metabolismo , Activación Transcripcional
9.
Toxicol Mech Methods ; 23(4): 217-22, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23193996

RESUMEN

Aging is attributed to both genetic and environmental factors. Occupational exposure is one of the environmental factors with potential genotoxic effects. Researchers try to determine factors involved in genetic damages at hazards exposure that could accelerate aging. Cytochrome P450 2E1 (CYP2E1) gene contributes in activation and detoxification of the environmental hazards. This polymorphism plays an important role in susceptibility of inter-individuals to DNA damage at the occupational exposure. The current study evaluated the possible influence of this gene polymorphism in aging by genomic damages through the biomarkers alterations of micronuclei (MN), comet tail length and telomere length shortening at the exposure. In this study, buccal cells were collected from the oral cavity of exposed workers and non-exposed controls. The CYP2E1 genotypes were detected by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). The wild genotype significantly affected MN frequency (p = 0.007) and relative telomere length (p = 0.047) in the older group of workers. It was concluded that the interaction of gene polymorphism and exposure enhances DNA damage and accelerates aging consequently.


Asunto(s)
Envejecimiento Prematuro , Automóviles , Citocromo P-450 CYP2E1/genética , Interacción Gen-Ambiente , Exposición Profesional/efectos adversos , Polimorfismo de Longitud del Fragmento de Restricción , Envejecimiento Prematuro/inducido químicamente , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Estudios de Casos y Controles , Ensayo Cometa , Daño del ADN , Humanos , Micronúcleos con Defecto Cromosómico/inducido químicamente , Pruebas de Micronúcleos , Mucosa Bucal/citología , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/enzimología , Reacción en Cadena en Tiempo Real de la Polimerasa , Riesgo , Factores Socioeconómicos , Acortamiento del Telómero/efectos de los fármacos , Acortamiento del Telómero/genética
10.
Aging Cell ; 11(4): 704-13, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22621437

RESUMEN

We undertook genetic and nongenetic approaches to investigate the relationship between telomere maintenance and osteoblast differentiation, as well as to uncover a possible link between a known mediator of cellular aging and senile bone loss. Using mouse models of disrupted telomere maintenance molecules, including mutants in the Werner helicase (Wrn(-/-) ), telomerase (Terc(-/-) ), and Wrn(-/-) Terc(-/-) double mutants predisposed to accelerated bone loss, we measured telomere dysfunction-induced foci (TIFs) and markers of osteoblast differentiation in mesenchymal progenitor cells (MPCs). We found that telomere maintenance is directly and significantly related to osteoblast differentiation, with dysfunctional telomeres associated with impaired differentiation independent of proliferation state. Telomere-mediated defects in osteoblast differentiation are associated with increased p53/p21 expression and concomitant reduction in RUNX2. Conversely, MPCs from p53(-/-) mice do not have substantial telomere dysfunction and spontaneously differentiate into osteoblasts. These results suggest that critical telomere dysfunction may be a prominent mechanism for age-related osteoporosis and limits MPC differentiation into bone-forming cells via the p53/p21 pathway.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento Prematuro/patología , Osteoblastos/patología , Homeostasis del Telómero/genética , Envejecimiento Prematuro/enzimología , Animales , Diferenciación Celular/genética , Senescencia Celular/genética , Senescencia Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Masculino , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/enzimología , Osteoporosis/enzimología , Osteoporosis/genética , Osteoporosis/patología , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Transducción de Señal , Telomerasa/deficiencia , Telomerasa/genética , Helicasa del Síndrome de Werner
11.
Aging Cell ; 11(3): 482-90, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22364217

RESUMEN

Telomerase is a ribonucleoprotein reverse transcriptase (RT) that synthesizes specific DNA repeats, or telomeric DNA, at the ends of chromosomes. Telomerase is minimally composed of a protein subunit, TERT, and an RNA component, TR. Aberrant telomerase activity has been associated with most human cancers and several premature aging diseases, such as idiopathic pulmonary fibrosis (IPF), a chronic, progressive, and fatal lung disease characterized by alveolar epithelial cell damage and fibrosis. Our study focuses on three hTERT mutations that were identified in a subset of patients with IPF, in which these patients also exhibited shorter telomeres compared with age-matched controls. We characterized how three IPF-associated hTERT mutations, V144M, R865C, and R865H, affected telomerase function both in vitro and in human cells. We demonstrated that the R865 residue is crucial for repeat addition processivity and thus telomere synthesis in telomerase-positive 293 cells and telomerase-negative BJ cells, consistent with its location in the hTERT nucleotide-binding motif. In contrast, while the V144M mutant did not exhibit any biochemical defects, this mutant was unable to elongate telomeres in human cells. As a result, our studies have identified hTERT V144 and R865 as two critical residues required for proper telomerase function in cells. Together, this may explain how inherited hTERT mutations can lead to shortened telomeres in patients with IPF and, thus, provide further insight into the role of naturally occurring telomerase mutations in the pathophysiology of certain age-related disease states.


Asunto(s)
Fibrosis Pulmonar Idiopática/enzimología , Fibrosis Pulmonar Idiopática/genética , Mutación , Telomerasa/genética , Telomerasa/metabolismo , Anciano de 80 o más Años , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Secuencia de Aminoácidos , Procesos de Crecimiento Celular/fisiología , Línea Celular , Fibroblastos/citología , Humanos , Fibrosis Pulmonar Idiopática/patología , Riñón/citología , Datos de Secuencia Molecular
12.
Proc Natl Acad Sci U S A ; 107(44): 18927-32, 2010 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-20956318

RESUMEN

Studies of long-lived Caenorhabditis elegans mutants have identified several genes that function to limit lifespan, i.e., loss-of-function mutations in these genes promote longevity. By contrast, little is known about genes that normally act to delay aging and that when mutated cause premature aging (progeria). To seek such genes, we performed a genetic screen for C. elegans mutants that age prematurely. We found that loss-of-function mutations of the ketoacyl thiolase gene kat-1 result in an increased accumulation of the lipofuscin-like fluorescent aging pigment, shortened lifespan, early behavioral decline, and other abnormalities characteristic of premature aging. These findings suggest that kat-1 acts to delay C. elegans aging. kat-1 encodes a conserved metabolic enzyme that catalyzes the last step of fatty acid oxidation and was previously shown to regulate fat accumulation in worms. We observed that kat-1 is required for the extension of lifespan and enhanced thermotolerance mediated by extra copies of the deacetylase gene sir-2.1. kat-1 acts independently of other known pathways that affect longevity. Our findings suggest that defects in fatty acid oxidation can limit lifespan and accelerate aging in C. elegans and that kat-1-mediated fatty acid oxidation is crucial for overexpressed sir-2.1 to delay aging.


Asunto(s)
Acetil-CoA C-Aciltransferasa/metabolismo , Envejecimiento/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Ácidos Grasos/metabolismo , Sirtuinas/metabolismo , Acetil-CoA C-Aciltransferasa/genética , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Ácidos Grasos/genética , Lipofuscina/genética , Lipofuscina/metabolismo , Mutación , Oxidación-Reducción , Progeria/enzimología , Progeria/genética , Sirtuinas/genética
13.
Indian J Exp Biol ; 48(4): 378-82, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20726336

RESUMEN

Effect of hydroalcoholic extract T. bispinosa (TB) was studied on fluorescence product and biochemical parameter like lipid peroxidation, catalase activity and glutathione peroxidase activity in the brain of female albino mice. Ageing was accelerated by the treatment of 0.5 ml 5% D-galactose for 15 days. This resulted in increased fluorescence product, increase lipid peroxidation and decrease antioxidant enzyme like glutathione peroxides and catalase in cerebral cortex. After cotreatment with hydroalcoholic extract of TB (500 mg/kg, po) there was decrease in fluorescence product in cerebral cortex. Moreover, TB inhibited increase lipid peroxidation and restores glutathione peroxidase and catalase activity in cerebral cortex as compare to ageing accelerated control group. To conclude TB found to be effective antioxidative agent which could to some extent reverse D-galactose induced ageing changes resulted due to oxidative damage.


Asunto(s)
Envejecimiento Prematuro/prevención & control , Encéfalo/efectos de los fármacos , Galactosa/toxicidad , Lipofuscina/biosíntesis , Lythraceae/química , Fármacos Neuroprotectores/farmacología , Extractos Vegetales/farmacología , Envejecimiento Prematuro/inducido químicamente , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Catalasa/metabolismo , Modelos Animales de Enfermedad , Femenino , Frutas/química , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Ratones , Fármacos Neuroprotectores/aislamiento & purificación , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/aislamiento & purificación
14.
Gerontology ; 56(4): 390-403, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20016137

RESUMEN

A defining feature of the eukaryotic genome is the presence of linear chromosomes. This arrangement, however, poses several challenges with regard to chromosomal replication and maintenance. To prevent the loss of coding sequences and to suppress gross chromosomal rearrangements, linear chromosomes are capped by repetitive nucleoprotein structures, called telomeres. Each cell division results in a progressive shortening of telomeres that, below a certain threshold, promotes genome instability, senescence, and apoptosis. Telomeric erosion, maintenance, and repair take center stage in determining cell fate. Cells of the immune system are under enormous proliferative demand, stressing telomeric intactness. Lymphocytes are capable of upregulating telomerase, an enzyme that can elongate telomeric sequences and, thus, prolong cellular lifespan. Therefore, telomere dynamics are critical in preserving immune function and have become a focus for studies of immunosenescence and autoimmunity. In this review, we describe the role of telomeric nucleoproteins in shaping telomere architecture and in suppressing DNA damage responses. We summarize new insights into the regulation of telomerase activity, hereditary disorders associated with telomere dysfunction, the role of telomere loss in immune aging, and the impact of telomere dysfunction in chronic inflammatory disease.


Asunto(s)
Envejecimiento/genética , Envejecimiento/inmunología , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/inmunología , Telómero/genética , Telómero/inmunología , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento Prematuro/enzimología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/inmunología , Animales , Senescencia Celular , Regulación Enzimológica de la Expresión Génica , Humanos , Enfermedades del Sistema Inmune/enzimología , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Modelos Biológicos , Linfocitos T/enzimología , Linfocitos T/inmunología , Linfocitos T/patología , Telomerasa/genética , Telomerasa/metabolismo , Telómero/enzimología
15.
Aging Cell ; 8(4): 502-6, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19416127

RESUMEN

Deletions in mitochondrial DNA (mtDNA) have long been suspected to be involved in mammalian aging, but their role remains controversial. Recent research has demonstrated that relatively higher levels of mtDNA deletions correlate with premature aging in mtDNA mutator mice, which led to the conclusion that premature aging in these mice is driven by mtDNA deletions. However, it is reported here that the absolute level of deletions in mutator mice is quite low, especially when compared with the level of point mutations in these mice. It is thus argued that the available data are insufficient to conclude that mtDNA mutations drive premature aging in mtDNA mutator mice. It remains possible that clonal expansion of mtDNA deletions may result in sufficiently high levels to play a role in age-related dysfunction in some cells, but assessing this possibility will require studies of the distribution of these deletions among different cell types and in individual cells.


Asunto(s)
Envejecimiento Prematuro/genética , ADN Mitocondrial/genética , Eliminación de Gen , Mutación , Envejecimiento Prematuro/enzimología , Animales , Encéfalo/metabolismo , ADN Polimerasa gamma , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Ratones , Ratones Transgénicos , Fenotipo
16.
Biochem Biophys Res Commun ; 383(3): 358-62, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-19364500

RESUMEN

Akt/PKB plays a pivotal role in cell survival and proliferation. Previously, we reported that UV-irradiation induces extensive cell death in Akt2(-/-) mouse embryonic fibroblasts (MEFs) while Akt1(-/-) MEFs show cell cycle arrest. Here, we find that Akt1(-/-) MEFs exhibit phenotypic changes characteristics of senescence upon UV-irradiation. An enlarged and flattened morphology, a reduced cell proliferation and an increased senescence-associated beta-galactosidase (SA beta-gal) staining indicate that Akt1(-/-) MEFs undergo premature senescence after UV-irradiation. Restoring Akt1 expression in Akt1(-/-) MEFs suppressed SA beta-gal activity, indicating that UV-induced senescence is due to the absence of Akt1 function. Notably, levels of ROS were rapidly increased upon UV-irradiation and the ROS scavenger NAC inhibits UV-induced senescence of Akt1(-/-) MEFs, suggesting that UV light induces premature senescence in Akt1(-/-) MEFs by modulating intracellular levels of ROS. In conjunction with our previous work, this indicates that different isoforms of Akt have distinct function in response to UV-irradiation.


Asunto(s)
Envejecimiento Prematuro/enzimología , Senescencia Celular/genética , Fibroblastos/efectos de la radiación , Proteínas Proto-Oncogénicas c-akt/genética , Especies Reactivas de Oxígeno/metabolismo , Rayos Ultravioleta/efectos adversos , Envejecimiento Prematuro/genética , Animales , Línea Celular , Senescencia Celular/efectos de los fármacos , Fibroblastos/enzimología , Ratones
17.
Biochem Biophys Res Commun ; 378(1): 103-7, 2009 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-19013131

RESUMEN

Aging is the greatest risk factor for neurodegenerative diseases such as Alzheimer's disease (AD). Age-dependent alterations of cell signaling play an important role in the onset of AD. The serine/threonine kinase Akt is a critical cell signaling to neuronal survival. Using the senescence-accelerated mouse SAMP10, we investigated the effect of aging on AKT signaling in hippocampus tissue. During aging, the expression of Akt mRNA and protein remained stable. However, the constructive phosphorylation of Akt(Ser473) displayed a continuous decrease after 6 months in SAMP10. When compared with the control SAMR1, aged SAMP10 mice showed significant reduced phosphorylation of Akt(Ser473). SAMP10 at the age of 6 months showed obvious deterioration in performance of learning and memory tasks. Thus, the data reported here suggested a potential link between the age-related alteration of Akt(Ser473) and the deterioration in performance of learning and memory tasks in SAMP10 mouse.


Asunto(s)
Envejecimiento Prematuro/enzimología , Envejecimiento/metabolismo , Enfermedad de Alzheimer/enzimología , Hipocampo/enzimología , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Factores de Edad , Enfermedad de Alzheimer/fisiopatología , Animales , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Mutantes , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Serina/metabolismo , Transducción de Señal
18.
Biogerontology ; 10(3): 253-66, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18830681

RESUMEN

The premature ageing ataxia telangiectasia (AT) and Werner syndromes (WS) are associated with accelerated cellular ageing. Young WS fibroblasts have an aged appearance and activated p38 MAP kinase, and treatment with the p38 inhibitor SB230580 extends their lifespan to within the normal range. SB203580 also extends the replicative lifespan of normal adult dermal fibroblasts, however, the effect is much reduced when compared to WS cells, suggesting that WS fibroblasts undergo a form of stress-induced premature senescence (SIPS). A small lifespan extension is seen in AT cells, which is not significant compared to normal fibroblasts, and the majority of young AT cells do not have an aged appearance and lack p38 activation, suggesting that the premature ageing does not result from SIPS. The lack of p38 activation is supported by the clinical manifestation, since AT is not associated with inflammatory disease, whereas WS individuals are predisposed to atherosclerosis, type II diabetes and osteoporosis, conditions known to be associated with p38 activation.


Asunto(s)
Envejecimiento Prematuro/patología , Ataxia Telangiectasia/patología , Senescencia Celular , Fibroblastos/patología , Transducción de Señal , Estrés Fisiológico , Síndrome de Werner/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Envejecimiento Prematuro/enzimología , Ataxia Telangiectasia/enzimología , Proliferación Celular , Forma de la Célula , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Activación Enzimática , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Síndrome de Werner/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
19.
Aging (Albany NY) ; 1(3): 289-302, 2009 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-20157518

RESUMEN

Werner syndrome is a premature aging disease caused by loss of function mutations in the Werner syndrome protein (WRN) gene. WRN is a RecQ helicase that in contrast to every other member of this family of proteins possesses an exonuclease activity. The findings that cells lacking WRN activity display accelerated telomere shortening and WRN can be detected at chromosome ends suggest that this protein participates in some aspects of telomere metabolism. In this study we examined the impact of WRN on telomeric substrates with a 3' single-stranded overhang in vitro and show that WRN has sequence-specific exonuclease activity that removes several nucleotides inward with a periodical pattern from the 3' end of the telomeric overhang. This activity is strictly dependent on the presence of telomeric sequences in both the duplex DNA and 3' overhang DNA segment and is strongly inhibited by the telomeric factor POT1 but not TRF2. These data demonstrate that WRN processes telomeric DNA substrates with a 3' single-stranded overhang with high specificity and suggest that this protein could influence the configuration of telomere ends prior to the formation of a protective t-loop structure.


Asunto(s)
Exodesoxirribonucleasas/metabolismo , RecQ Helicasas/metabolismo , Telómero , Envejecimiento Prematuro/enzimología , Composición de Base , Secuencia de Bases , ADN de Cadena Simple , Exodesoxirribonucleasas/química , Homeostasis , Humanos , RecQ Helicasas/química , Complejo Shelterina , Especificidad por Sustrato , Telómero/química , Telómero/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Factores de Tiempo , Síndrome de Werner/enzimología , Síndrome de Werner/patología , Helicasa del Síndrome de Werner
20.
Mol Cell Proteomics ; 7(9): 1737-47, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18515266

RESUMEN

The liver is a complex and unique organ responsible for a breadth of functions crucial to sustaining life, especially for various metabolic processes in its mitochondria. Senescence-accelerated mouse prone/8 (SAMP8), a widely used aging model, exhibits an oxidative stress-induced aging phenotype and severe mitochondria-related liver pathology that are not seen in senescence-accelerated mouse resistant/1 (SAMR1). Here we used both two-dimensional electrophoresis- and ICAT-based mitochondrial proteomics analysis to view the liver mitochondrial protein alterations between SAMP8 and SAMR1. Compared with SAMR1, decreased expression and activity of mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase were detected in SAMP8 at 6 months old (SAMP8-6m). As the key enzyme of ketogenesis, 3-hydroxy-3-methylglutaryl-CoA synthase is well known to be transcriptionally regulated by peroxisome proliferator-activated receptor alpha, which was also expressed at lower levels in SAMP8-6m livers. In addition, down-regulation of two peroxisome proliferator-activated receptor alpha target gene products (acyl-CoA oxidase and enoyl-CoA hydratase), elevation of triglyceride, and reduction of acetyl-CoA were observed, indicating abnormal fatty acid metabolism in SAMP8-6m livers. In addition eight proteins (NDUAA, NDUBA, NDUB7, NDUS1, NDUS3, NDUV1, ETFA, and UCRI) of mitochondrial complexes were down-regulated in SAMP8-6m, resulting in mitochondria-related liver dysfunction characterized by enhanced oxidative stress-induced molecular damage (lipid peroxide and oxidized protein) and depressed energy production (ATP). Glutamine synthetase and ornithine aminotransferase involved in glutamine synthesis were up-regulated in SAMP8 livers at both 1 and 6 months old that may be related to the accumulation of glutamate and glutamine. Our work provided useful clues to understanding the molecular mechanism underlying liver dysfunction in senescence-accelerated mouse.


Asunto(s)
Envejecimiento Prematuro/enzimología , Hígado/enzimología , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Proteómica/métodos , Adenosina Trifosfato/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/fisiopatología , Animales , Modelos Animales de Enfermedad , Electroforesis en Gel Bidimensional , Hígado/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Proteínas Mitocondriales/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA