Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Biomed Pharmacother ; 171: 116110, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38198955

RESUMEN

Skin is susceptible to premature aging in response to ultraviolet (UV) radiation-induced oxidative stress, which can ultimately result in aberrant aging or age-related disorders. Accordingly, strategies that can be adopted to mitigate oxidative stress may contribute to protecting skin from induced aging-related damage, thereby offering promising approaches for the treatment of skin diseases and disorders. In this regard, oroxylin A (OA), a natural flavonoid isolated from certain plants used in traditional Chinese medicine, is considered to have notable antioxidant, anti-inflammatory, and anti-apoptotic properties, and is often used to treat certain inflammatory diseases. To date, however, there has been comparatively little research on the effects of OA with respect skin aging. In this study, we utilized UV radiation-induced mouse and cellular models of aging to assess the efficacy of OA in protecting against skin aging. Subsequently, to elucidate the potential mechanisms underlying the protective effect of OA on skin aging, we performed molecular docking analysis to investigate the involvement of the anti-aging gene Sirt1, which was further confirmed on the basis of Sirt1 gene silencing. We accordingly demonstrated that by promoting an increase in the expression of Sirt1, OA can contribute to suppressing UV-induced skin photo-aging in cells/mice by reducing oxidative stress. Furthermore, we established that by activating Sirt1, OA can also promote the dissociation of Nrf2 from Keap1 and its subsequent nuclear translocation. Collectively, our findings in this study reveal OA to be an effective natural compound that can be administered to delay the aging of skin triggered by UV, both in vivo and in vitro, by binding to Sirt1 to promote the deacetylation and nuclear translocation of Nrf2, thereby contributing to a reduction in oxidative stress. These findings may this provide a therapeutic target for the prevention of skin aging or aging-induced skin diseases.


Asunto(s)
Envejecimiento Prematuro , Flavonoides , Envejecimiento de la Piel , Enfermedades de la Piel , Animales , Ratones , Envejecimiento Prematuro/tratamiento farmacológico , Flavonoides/farmacología , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Simulación del Acoplamiento Molecular , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Sirtuina 1/metabolismo , Envejecimiento de la Piel/efectos de los fármacos , Enfermedades de la Piel/tratamiento farmacológico , Rayos Ultravioleta
2.
Aging Cell ; 22(12): e13983, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37858983

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal genetic condition that arises from a single nucleotide alteration in the LMNA gene, leading to the production of a defective lamin A protein known as progerin. The accumulation of progerin accelerates the onset of a dramatic premature aging phenotype in children with HGPS, characterized by low body weight, lipodystrophy, metabolic dysfunction, skin, and musculoskeletal age-related dysfunctions. In most cases, these children die of age-related cardiovascular dysfunction by their early teenage years. The absence of effective treatments for HGPS underscores the critical need to explore novel safe therapeutic strategies. In this study, we show that treatment with the hormone ghrelin increases autophagy, decreases progerin levels, and alleviates other cellular hallmarks of premature aging in human HGPS fibroblasts. Additionally, using a HGPS mouse model (LmnaG609G/G609G mice), we demonstrate that ghrelin administration effectively rescues molecular and histopathological progeroid features, prevents progressive weight loss in later stages, reverses the lipodystrophic phenotype, and extends lifespan of these short-lived mice. Therefore, our findings uncover the potential of modulating ghrelin signaling offers new treatment targets and translational approaches that may improve outcomes and enhance the quality of life for patients with HGPS and other age-related pathologies.


Asunto(s)
Envejecimiento Prematuro , Progeria , Adolescente , Niño , Humanos , Ratones , Animales , Progeria/tratamiento farmacológico , Progeria/genética , Progeria/metabolismo , Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/genética , Ghrelina/farmacología , Calidad de Vida , Piel/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Envejecimiento
3.
Nat Aging ; 3(2): 185-201, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-37118121

RESUMEN

An accumulating body of evidence indicates an association between mitotic defects and the aging process in Hutchinson-Gilford progeria syndrome (HGPS), which is a premature aging disease caused by progerin accumulation. Here, we found that BUBR1, a core component of the spindle assembly checkpoint, was downregulated during HGPS cellular senescence. The remaining BUBR1 was anchored to the nuclear membrane by binding with the C terminus of progerin, thus further limiting the function of BUBR1. Based on this, we established a unique progerin C-terminal peptide (UPCP) that effectively blocked the binding of progerin and BUBR1 and enhanced the expression of BUBR1 by interfering with the interaction between PTBP1 and progerin. Finally, UPCP significantly inhibited HGPS cellular senescence and ameliorated progeroid phenotypes, extending the lifespan of LmnaG609G/G609G mice. Our findings reveal an essential role for the progerin-PTBP1-BUBR1 axis in HGPS. Therapeutics designed around UPCP may be a beneficial strategy for HGPS treatment.


Asunto(s)
Envejecimiento Prematuro , Progeria , Ratones , Animales , Progeria/tratamiento farmacológico , Envejecimiento Prematuro/tratamiento farmacológico , Fenotipo
4.
Oxid Med Cell Longev ; 2021: 9942090, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34413931

RESUMEN

The roots of Vicatia thibetica de Boiss are a kind of Chinese herb with homology of medicine and food. This is the first report showing the property of the extract of Vicatia thibetica de Boiss roots (HLB01) to extend the lifespan as well as promote the healthy parameters in Caenorhabditis elegans (C. elegans). For doxorubicin- (Doxo-) induced premature aging in adult mice, HLB01 counteracted the senescence-associated biomarkers, including P21 and γH2AX. Interestingly, HLB01 promoted the expression of collagen in C. elegans and mammalian cell systemically, which might be one of the essential factors to exert the antiaging effects. In addition, HLB01 was also found as a scavenger of free radicals, thereby performing the antioxidant ability. Lifespan extension by HLB01 was also dependent on DAF-16 and HSF-1 via oxidative stress resistance and heat stress resistance. Taken together, overall data suggested that HLB01 could extend the lifespan and healthspan of C. elegans and resist Doxo-induced senescence in mice via promoting the expression of collagen, antioxidant potential, and stress resistance.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Antioxidantes/farmacología , Apiaceae/química , Caenorhabditis elegans/crecimiento & desarrollo , Doxorrubicina/toxicidad , Longevidad , Extractos Vegetales/farmacología , Envejecimiento Prematuro/inducido químicamente , Envejecimiento Prematuro/patología , Animales , Antibióticos Antineoplásicos/toxicidad , Caenorhabditis elegans/efectos de los fármacos , Respuesta al Choque Térmico , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Raíces de Plantas/química
5.
Transl Neurodegener ; 10(1): 12, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789744

RESUMEN

BACKGROUND: Currently, there is no cure for Alzheimer's disease (AD). Therapeutics that can modify the early stage of AD are urgently needed. Recent studies have shown that the pathogenesis of AD is closely regulated by an endo/lysosomal asparaginyl endopeptidase (AEP). Inhibition of AEP has been reported to prevent neural degeneration in transgenic mouse models of AD. However, more than 90% of AD cases are age-related sporadic AD rather than hereditary AD. The therapeutic efficacy of AEP inhibition in ageing-associated sporadic AD remains unknown. METHODS: The senescence-accelerated mouse prone 8 (SAMP8) was chosen as an approximate model of sporadic AD and treated with a selective AEP inhibitor,: δ-secretase inhibitor 11. Activation of AEP was determined by enzymatic activity assay. Concentration of soluble amyloid ß (Aß) in the brain was determined by ELISA. Morris water maze test was performed to assess the learning and memory-related cognitive ability. Pathological changes in the brain were explored by morphological and western blot analyses. RESULTS: The enzymatic activity of AEP in the SAMP8 mouse brain was significantly higher than that in the age-matched SAMR1 mice. The half maximal inhibitory concentration (IC50) for δ-secretase inhibitor 11 to inhibit AEP in vitro is was around 150 nM. Chronic treatment with δ-secretase inhibitor 11 markedly decreased the brain AEP activity, reduced the generation of Aß1-40/42 and ameliorated memory loss. The inhibition of AEP with this reagent not only reduced the AEP-cleaved tau fragments and tau hyperphosphorylation, but also attenuated neuroinflammation in the form of microglial activation. Moreover, treatment with δ-secretase inhibitor 11 prevented the synaptic loss and alleviated dendritic disruption in SAMP8 mouse brain. CONCLUSIONS: Pharmacological inhibition of AEP can intervene and prevent AD-like pathological progress in the model of sporadic AD. The up-regulated AEP in the brain could be a promising target for early treatment of AD. The δ-secretase inhibitor 11 can be used as a lead compound for translational development of AD treatment.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/patología , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Cisteína Endopeptidasas/metabolismo , Inhibidores de Proteasas/uso terapéutico , Envejecimiento , Péptidos beta-Amiloides/análisis , Animales , Encéfalo/patología , Química Encefálica , Cognición , Cisteína Endopeptidasas/efectos de los fármacos , Humanos , Masculino , Aprendizaje por Laberinto , Memoria , Ratones
6.
Immunobiology ; 225(1): 151870, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31822433

RESUMEN

Senescence is an inevitable and complicated phenomenon. Age-associated thymic involution increases the risk of infectious diseases, which results in the immunosenescence and leads to a poor immune function. d-galactose (d-gal) can cause damages that resemble accelerated aging in mice. Gallic acid (GA), as one of the natural phenolic compounds, has been demonstrated to act in antioxidant and anti-tumor effects. In this study, we explored the effects of GA in preventing the age-related thymic involution and the alterations of the forkhead box protein N1 (FoxN1) in d-gal induced accelerated aging mice. The accelerated aging mice model was established by intraperitoneal injection d-gal for eight weeks and given GA with 200, 250, 500 mg/kg body weight per day, respectively, for six weeks. It showed that the d-gal-treated mice developed structural changes in the thymi compared to normal control mice. With supplement of GA, the mice restored the normal thymic anatomy, including the thickening cortex compartment and clearer cortico-medullary junction. The d-gal-treated mice showed a severe reduction in the number of thymocytes, GA mice also displayed the increased numbers of CD4 + T cells through flow cytometric analysis. GA treatment increased the proliferative cells by BrdU incorporation assay and reduced the numbers of apoptotic cells with FITC-12-dUTP labeling (TUNEL). The expression of FoxN1 was also found increased in GA treated mice by immunohistochemistry and quantitative reverse transcriptase PCR (qRT-PCR). Taken together, our results suggested that the administration of GA opposed the involution of thymus via stimulation of FoxN1 expression and proliferation of cells in a dose-dependent manner.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Linfocitos T CD4-Positivos/patología , Factores de Transcripción Forkhead/metabolismo , Ácido Gálico/uso terapéutico , Timocitos/patología , Timo/anatomía & histología , Envejecimiento Prematuro/inducido químicamente , Animales , Recuento de Células , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Galactosa , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos ICR , Tamaño de los Órganos , Timo/efectos de los fármacos
7.
Med Hypotheses ; 134: 109406, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31634774

RESUMEN

We introduce a novel hypothesis which states that the therapeutic utilisation of psilocybin has beneficial effects on genetic aging. Ex hypothesi, we predict a priori that controlled psilocybin interventions exert quantifiable positive impact on leucocyte telomere length (telomeres are a robust predictor of mortality and multifarious aging-related diseases). Our hypothesising follows the Popperian logic of scientific discovery, viz., bold (and refutable) conjectures form the very foundation of scientific progress. The 'psilocybin-telomere hypothesis' is formalised as a logically valid deductive (syllogistic) argument and we provide substantial evidence to support the underlying premises. Impetus for our theorising derives from a plurality of converging empirical sources indicating that psilocybin has persistent beneficial effects on various aspects of mental health (e.g., in the context of depression, anxiety, PTSD, OCD, addiction, etc.). Additional support is based on a large corpus of studies that establish reliable correlations between mental health and telomere attrition (improved mental health is generally correlated with longer telomeres). Another pertinent component of our argument is based on recent studies which demonstrate that "meditative states of consciousness" provide beneficial effects on genetic aging. Similarly, psilocybin can induce states of consciousness that are neurophysiologically and phenomenologically significantly congruent with meditative states. Furthermore, prior research has demonstrated that a single dose of psilocybin can occasion life-changing transformative experiences (≈ 70% of healthy volunteers rate their experience with psilocybin amongst the five personally most meaningful lifetime events, viz., ranked next to giving birth to a child or losing a loved one). We postulate that these profound psychological events leave quantifiable marks at the molecular genetic/epigenetic level. Given the ubiquitous availability and cost effectiveness of telomere length assays, we suggest that quantitative telomere analysis should be regularly included in future psilocybin studies as an adjunctive biological marker (i.e., to facilitate scientific consilience via methodological triangulation). In order to substantiate the 'psilocybin-telomere hypothesis' potential neuropsychopharmacological, endocrinological, and genetic mechanisms of action are discussed (e.g., HPA-axis reactivity, hippocampal neurogenesis, neurotropic growth factors such as BDNF, 5-HT2A receptor agonism, neuroplasticity/synaptoplasticity, brain-wide alterations in neuronal functional connectivity density, involvement of the SLC6A4 serotonin transporter gene, inter alia). The proposed research agenda is thus intrinsically highly interdisciplinary, and it has deep ramifications from a philosophy of science perspective as it connects the epistemic level (qualitative experiential phenomenology) with the ontic level (quantitative molecular genetics) of analysis. In the long term, multidisciplinary and innovative investigations of the 'psilocybin-telomere hypothesis' could contribute to the improvement of senotherapeutic psychological interventions and the identification of novel geroprotective and neuroprotective/restorative pharmaceutical targets to decelerate genetic aging and improve well-being and quality of life during the aging process.


Asunto(s)
Envejecimiento/efectos de los fármacos , Modelos Genéticos , Modelos Psicológicos , Psilocibina/uso terapéutico , Psicotrópicos/uso terapéutico , Acortamiento del Telómero/efectos de los fármacos , Envejecimiento/genética , Envejecimiento/psicología , Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/prevención & control , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Factor Neurotrófico Derivado del Encéfalo/fisiología , Estado de Conciencia/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Depresión/tratamiento farmacológico , Depresión/genética , Modelos Animales de Enfermedad , Sistema Endocrino/fisiopatología , Humanos , Neurotransmisores/fisiología , Estrés Oxidativo/efectos de los fármacos , Personalidad/efectos de los fármacos , Psilocibina/farmacología , Psicotrópicos/farmacología , Proyectos de Investigación , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/genética , Acortamiento del Telómero/fisiología
8.
Int J Med Mushrooms ; 21(4): 381-392, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31002633

RESUMEN

We studied Phellinus lonicerinus to determine the cytotoxic effect and the dual estrogenic activities of methyl-hispolon and their relation to estrogen signals in vivo and in vitro. The Glide scores of methyl-hispolon-estrogen receptor α (ERα) and methyl-hispolon-ERß docked complexes were -7.29 kcal/mol and -6.68 kcal/mol in docking simulations. Methyl-hispolon had a significant antiproliferative effect for estrogen-sensitive ER(+) MCF-7 cells in the absence of estrogen, and it exhibited dual estrogen activities. Methyl-hispolon increased the serum E2 in rats with premature ovarian failure and fulfilled the estrogenic function in the uterus and ovary. Methyl-hispolon significantly inhibited the expression of Ras, API, ERα, C-myc, and cyclinDl, as well as their gene transcription in RL95-2 cells. The phosphorylation of ERK1/2 was inhibited by methyl-hispolon. Thus, methyl-hispolon has potential use in treating estrogen deficiency-related diseases, with good antitumor effects and estrogenic activity.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Basidiomycota/química , Catecoles/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Estrógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Catecoles/química , Proliferación Celular/efectos de los fármacos , Moduladores de los Receptores de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Terapia de Reemplazo de Hormonas , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Ovario/efectos de los fármacos , Fosforilación , Fitoestrógenos/metabolismo , Ratas , Ratas Sprague-Dawley , Útero/efectos de los fármacos
9.
Protein Cell ; 10(6): 417-435, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30069858

RESUMEN

Aging increases the risk of various diseases. The main goal of aging research is to find therapies that attenuate aging and alleviate aging-related diseases. In this study, we screened a natural product library for geroprotective compounds using Werner syndrome (WS) human mesenchymal stem cells (hMSCs), a premature aging model that we recently established. Ten candidate compounds were identified and quercetin was investigated in detail due to its leading effects. Mechanistic studies revealed that quercetin alleviated senescence via the enhancement of cell proliferation and restoration of heterochromatin architecture in WS hMSCs. RNA-sequencing analysis revealed the transcriptional commonalities and differences in the geroprotective effects by quercetin and Vitamin C. Besides WS hMSCs, quercetin also attenuated cellular senescence in Hutchinson-Gilford progeria syndrome (HGPS) and physiological-aging hMSCs. Taken together, our study identifies quercetin as a geroprotective agent against accelerated and natural aging in hMSCs, providing a potential therapeutic intervention for treating age-associated disorders.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Células Madre Mesenquimatosas , Progeria/tratamiento farmacológico , Quercetina/farmacología , Síndrome de Werner/tratamiento farmacológico , Envejecimiento/efectos de los fármacos , Ácido Ascórbico/farmacología , Línea Celular , Proliferación Celular , Senescencia Celular/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Modelos Biológicos
10.
Nat Commun ; 9(1): 1700, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29703891

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, but devastating genetic disease characterized by segmental premature aging, with cardiovascular disease being the main cause of death. Cells from HGPS patients accumulate progerin, a permanently farnesylated, toxic form of Lamin A, disrupting the nuclear shape and chromatin organization, leading to DNA-damage accumulation and senescence. Therapeutic approaches targeting farnesylation or aiming to reduce progerin levels have provided only partial health improvements. Recently, we identified Remodelin, a small-molecule agent that leads to amelioration of HGPS cellular defects through inhibition of the enzyme N-acetyltransferase 10 (NAT10). Here, we show the preclinical data demonstrating that targeting NAT10 in vivo, either via chemical inhibition or genetic depletion, significantly enhances the healthspan in a Lmna G609G HGPS mouse model. Collectively, the data provided here highlights NAT10 as a potential therapeutic target for HGPS.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Inestabilidad Genómica/efectos de los fármacos , Hidrazonas/farmacología , Acetiltransferasa A N-Terminal/antagonistas & inhibidores , Progeria/tratamiento farmacológico , Tiazoles/farmacología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/mortalidad , Envejecimiento Prematuro/patología , Animales , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Modelos Animales de Enfermedad , Femenino , Inestabilidad Genómica/genética , Humanos , Hidrazonas/uso terapéutico , Estimación de Kaplan-Meier , Lamina Tipo A/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Acetiltransferasa A N-Terminal/genética , Acetiltransferasa A N-Terminal/metabolismo , Acetiltransferasas N-Terminal , Progeria/genética , Progeria/mortalidad , Progeria/patología , Tiazoles/uso terapéutico
11.
Mol Med Rep ; 17(5): 6947-6960, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29568888

RESUMEN

The present study aimed to investigate the possible effects and underlying molecular mechanism of Bushen­Yizhi formula (BSYZ), a traditional Chinese medicine, on age­related degeneration of brain physiology in senescence­accelerated mouse prone 8 (SAMP8) mice. SAMP8 mice (age, 6 months) were administered BSYZ (1.46, 2.92 and 5.84 g/kg/day) for 30 days. Morris water maze and step­down tests demonstrated that BSYZ significantly improved memory impairments in SAMP8 mice. In addition, BSYZ significantly enhanced the expression levels of peroxisome proliferator­activated receptor­Î³ and B­cell lymphoma extra­large, and downregulated the expression levels of inflammatory mediators, glial fibrillary acidic protein, cyclooxygenase­2, nuclear factor­κB and interleukin­1ß in the brain compared with untreated SAMP8 mice. Furthermore, BSYZ reversed disordered superoxide dismutase activity, malondialdehyde content and glutathione peroxidase activity, and ameliorated apoptosis and histological alterations. The present study indicated that BSYZ may attenuate cognitive impairment in SAMP8 mice, and modulate inflammation, oxidative stress and neuronal apoptosis. These results suggested that BSYZ may have the potential to be further developed into a therapeutic agent for protection against age­related neurodegenerative diseases.


Asunto(s)
Envejecimiento Prematuro/complicaciones , Envejecimiento Prematuro/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Inflamación/tratamiento farmacológico , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Química Encefálica/efectos de los fármacos , Ciclooxigenasa 2/análisis , Proteína Ácida Fibrilar de la Glía/análisis , Inflamación/etiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , PPAR gamma/análisis
12.
Arch Dermatol Res ; 309(4): 275-283, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28247017

RESUMEN

Skin Aging manifests primarily with wrinkles, dyspigmentations, texture changes, and loss of elasticity. During the skin aging process, there is a loss of moisture and elasticity in skin resulting in loss of firmness finally leading to skin sagging. The key molecule involved in skin moisture is hyaluronic acid (HA), which has a significant water-binding capacity. HA levels in skin decline with age resulting in decrease in skin moisture, which may contribute to loss of firmness. Clinical trials have shown that topically applied ROL effectively reduces wrinkles and helps retain youthful appearance. In the current study, ROL was shown to induce HA production and stimulates the gene expression of all three forms of hyaluronic acid synthases (HAS) in normal human epidermal keratinocytes monolayer cultures. Moreover, in human skin equivalent tissues and in human skin explants, topical treatment of tissues with a stabilized-ROL formulation significantly induced the gene expression of HAS mRNA concomitant with an increased HA production. Finally, in a vehicle-controlled human clinical study, histochemical analysis confirmed increased HA accumulation in the epidermis in ROL-treated human skin as compared to vehicle. These results show that ROL increases skin expression of HA, a significant contributing factor responsible for wrinkle formation and skin moisture, which decrease during aging. Taken together with the activity to increase collagen, elastin, and cell proliferation, these studies establish that retinol provides multi-functional activity for photodamaged skin.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Glucuronosiltransferasa/metabolismo , Queratinocitos/efectos de los fármacos , Piel/efectos de los fármacos , Vitamina A/uso terapéutico , Administración Tópica , Células Cultivadas , Elastina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Humanos , Hialuronano Sintasas , Ácido Hialurónico/metabolismo , Queratinocitos/metabolismo , Técnicas de Cultivo de Órganos , Piel/patología , Envejecimiento de la Piel/efectos de los fármacos
13.
J Cardiovasc Pharmacol ; 68(5): 374-382, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27824722

RESUMEN

Bmal1-(brain and muscle ARNT-like protein-1) deficient (Bmal1) mice prematurely age because of an increased reactive oxygen species (ROS) production. These mice also show a decline in cardiac function with age. We investigated whether an antioxidant treatment can ameliorate the declining cardiac function in prematurely aged Bmal1 mice. Male Bmal1 and wild-type (Bmal1) mice were exposed for 15 weeks to a high fat and high cholesterol diet with or without the antioxidant 4-hydroxy-2,2,6,6-tetramethylpiperidin-1-oxyl (TEMPOL; 5 mmol/L; in drinking water during the last 10 weeks). Echocardiographic analysis revealed that TEMPOL treatment of Bmal1 mice normalized cardiac function, as evidenced by a decrease in left ventricular diastolic and systolic internal diameters, and by an increase in fractional shortening and ejection fraction. The antioxidant did not affect cardiac function in Bmal1 mice. Although TEMPOL did not influence cardiac ROS levels in Bmal1 mice, it significantly protected Bmal1 cardiac telomeres from oxidation, as evidenced by a reduction in the telomere damage score (0.11 ± 0.012% vs. 0.16 ± 0.015%; P = 0.028). Thus, antioxidant treatment normalized cardiac function of Bmal1 mice, probably in part by scavenging ROS.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/metabolismo , Antioxidantes/uso terapéutico , Óxidos N-Cíclicos/uso terapéutico , Modelos Animales de Enfermedad , Animales , Antioxidantes/farmacología , Colesterol en la Dieta/administración & dosificación , Colesterol en la Dieta/efectos adversos , Óxidos N-Cíclicos/farmacología , Dieta Alta en Grasa/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Marcadores de Spin , Resultado del Tratamiento
14.
Oncotarget ; 7(35): 55951-55956, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27507054

RESUMEN

GDF11 has recently emerged as a powerful anti-aging candidate, found in young blood, capable of rejuvenating a number of aged tissues, such as heart, skeletal muscle and brain. However, recent reports have shown contradictory data questioning its capacity to reverse age-related tissue dysfunction. The availability of a mouse model of accelerated aging, which shares most of the features occurring in physiological aging, gives us an excellent opportunity to test in vivo therapies aimed at extending lifespan both in pathological and normal aging. On this basis, we wondered whether the proposed anti-aging functions of GDF11 would have an overall effect on longevity. We first confirmed the existence of a reduction in GDF11/8 levels in our mouse model of accelerated aging compared with wild-type littermates. However, we show herein that GDF11 daily administration does not extend lifespan of premature-aged mice.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Proteínas Morfogenéticas Óseas/uso terapéutico , Factores de Diferenciación de Crecimiento/uso terapéutico , Longevidad/fisiología , Miostatina/metabolismo , Rejuvenecimiento/fisiología , Envejecimiento Prematuro/metabolismo , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Modelos Animales de Enfermedad , Femenino , Factores de Diferenciación de Crecimiento/metabolismo , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/uso terapéutico
15.
J Clin Endocrinol Metab ; 101(10): 3592-3599, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27459537

RESUMEN

CONTEXT: Type 2 Wolfram syndrome (T2-WFS) is a neuronal and ß-cell degenerative disorder caused by mutations in the CISD2 gene. The mechanisms underlying ß-cell dysfunction in T2-WFS are not known, and treatments that effectively improve diabetes in this context are lacking. OBJECTIVE: Unraveling the mechanisms of ß-cell dysfunction in T2-WFS and the effects of treatment with GLP-1 receptor agonist (GLP-1-RA). DESIGN AND SETTING: A case report and in vitro mechanistic studies. PATIENT AND METHODS: We treated an insulin-dependent T2-WFS patient with the GLP-1-RA exenatide for 9 weeks. An iv glucose/glucagon/arginine stimulation test was performed off-drug before and after intervention. We generated a cellular model of T2-WFS by shRNA knockdown of CISD2 (nutrient-deprivation autophagy factor-1 [NAF-1]) in rat insulinoma cells and studied the mechanisms of ß-cell dysfunction and the effects of GLP-1-RA. RESULTS: Treatment with exenatide resulted in a 70% reduction in daily insulin dose with improved glycemic control, as well as an off-drug 7-fold increase in maximal insulin secretion. NAF-1 repression in INS-1 cells decreased insulin content and glucose-stimulated insulin secretion, while maintaining the response to cAMP, and enhanced the accumulation of labile iron and reactive oxygen species in mitochondria. Remarkably, treatment with GLP-1-RA and/or the iron chelator deferiprone reversed these defects. CONCLUSION: NAF-1 deficiency leads to mitochondrial labile iron accumulation and oxidative stress, which may contribute to ß-cell dysfunction in T2-WFS. Treatment with GLP-1-RA and/or iron chelation improves mitochondrial function and restores ß-cell function. Treatment with GLP-1-RA, probably aided by iron chelation, should be considered in WFS and other forms of diabetes associated with iron dysregulation.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón/agonistas , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Enfermedades Mitocondriales/tratamiento farmacológico , Atrofia Óptica/tratamiento farmacológico , Péptidos/farmacología , Ponzoñas/farmacología , Animales , Exenatida , Femenino , Humanos , Hipoglucemiantes/administración & dosificación , Péptidos/administración & dosificación , Ratas , Ponzoñas/administración & dosificación
16.
Exp Gerontol ; 75: 1-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26656745

RESUMEN

Aging is associated with an increase in oxidative stress and inflammation. The aging lung is particularly affected since it is continuously exposed to environmental oxidants while antioxidant machinery weakens with age. Melatonin, a free radical scavenger, counteracts inflammation and apoptosis in healthy cells from several tissues. Its effects on the aging lung are, however, not yet fully understood. This study aimed to investigate the effect of chronic administration of melatonin on the expression of inflammation markers (TNF-α, IL-1ß, NFκB2, HO-1) and apoptosis parameters (BAD, BAX, AIF) in the lung tissue of male senescence-accelerated prone mice (SAMP8). In addition, RNA oxidative damage, as the formation of 8-hydroxyguanosine (8-OHG), was also evaluated. Young and old animals, aged 2 and 10 months respectively, were divided into 4 groups: untreated young, untreated old, old mice treated with 1mg/kg/day melatonin, and old animals treated with 10mg/kg/day melatonin. Untreated young and old male senescence accelerated resistant mice (SAMR1) were used as controls. After 30 days of treatment, animals were sacrificed. Lungs were collected and immediately frozen in liquid nitrogen. mRNA and protein expressions were measured by RT-PCR and Western blotting, respectively. Levels of 8-OHG were quantified by ELISA. Mean values were analyzed using ANOVA. Old nontreated SAMP8 animals showed increased (p<0.05) mRNA and protein levels of TNF-α, IL-1ß, NFκB2, and HO-1 compared to young mice and SAMR1 mice. Melatonin treatment with either dose reversed the aging-derived inflammation (p<0.05). BAD, BAX and AIF expressions also rose with aging, the effect being counteracted with melatonin (p<0.05). Aging also caused a significant elevation (p<0.05) in SAMP8 8-OHG values. This increase was not observed in animals treated with melatonin (p<0.05). In conclusion, melatonin treatment was able to modulate the inflammatory and apoptosis status of the aging lungs, exerting a protective effect on age-induced damage.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Melatonina/farmacología , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/patología , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Melatonina/administración & dosificación , Melatonina/uso terapéutico , Ratones Mutantes , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología
17.
Aging Cell ; 15(2): 279-90, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26663466

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS), a fatal premature aging disease, is caused by a single-nucleotide mutation in the LMNA gene. Previous reports have focused on nuclear phenotypes in HGPS cells, yet the potential contribution of the mitochondria, a key player in normal aging, remains unclear. Using high-resolution microscopy analysis, we demonstrated a significantly increased fraction of swollen and fragmented mitochondria and a marked reduction in mitochondrial mobility in HGPS fibroblast cells. Notably, the expression of PGC-1α, a central regulator of mitochondrial biogenesis, was inhibited by progerin. To rescue mitochondrial defects, we treated HGPS cells with a mitochondrial-targeting antioxidant methylene blue (MB). Our analysis indicated that MB treatment not only alleviated the mitochondrial defects but also rescued the hallmark nuclear abnormalities in HGPS cells. Additional analysis suggested that MB treatment released progerin from the nuclear membrane, rescued perinuclear heterochromatin loss and corrected misregulated gene expression in HGPS cells. Together, these results demonstrate a role of mitochondrial dysfunction in developing the premature aging phenotypes in HGPS cells and suggest MB as a promising therapeutic approach for HGPS.


Asunto(s)
Envejecimiento Prematuro/tratamiento farmacológico , Azul de Metileno/farmacología , Mitocondrias/efectos de los fármacos , Progeria/tratamiento farmacológico , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/patología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Progeria/genética , Progeria/metabolismo , Progeria/patología , Regulación hacia Arriba/efectos de los fármacos
18.
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
19.
Arch Dermatol Res ; 305(7): 595-602, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23912478

RESUMEN

The anti-skin aging effects of epigallocatechin-3-gallate (EGCG) have been studied extensively in vitro and in vivo models. Accumulating data suggest that EGCG possesses important antioxidant and photoprotective properties. Our previous study demonstrated that heat exposure induces cutaneous angiogenesis and inflammatory cellular infiltration, disrupts the dermal extracellular matrix by inducing matrix metalloproteinases, and alters dermal structural proteins, thereby causing premature skin aging. In the present study, we examined whether EGCG may inhibit expression of MMP-1 in heat-stimulated human dermal fibroblasts. Furthermore, we investigated the inhibitory mechanism of EGCG on heat-induced MMP-1 expression. Western blot analysis and MMP-1 promoter assay revealed that EGCG markedly inhibited heat shock-induced MMP-1 expression in human dermal fibroblasts. In addition, we found that heat shock increased AP-1 DNA binding activity, and c-Jun was found to be increased mostly by heat stimulation in a supershift assay, which were suppressed by EGCG treatment. Also, in Western blotting, EGCG significantly inhibited the heat-induced expression of AP-1 constituent proteins, c-Jun, JunB and c-Fos. These results demonstrated that EGCG has abilities to inhibit heat-induced collagenolytic MMP-1 production via interfering with AP-1 pathways. Therefore, we propose that EGCG may be a potential agent for the prevention and treatment for heat shock-induced skin aging (thermal skin aging).


Asunto(s)
Envejecimiento Prematuro/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Factor de Transcripción AP-1/metabolismo , Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/patología , Animales , Catequina/análogos & derivados , Catequina/farmacología , Dermis/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Calor/efectos adversos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , Metaloproteinasa 1 de la Matriz/genética , Ratones , Células 3T3 NIH , Unión Proteica , Transducción de Señal , Envejecimiento de la Piel/efectos de los fármacos , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/farmacología
20.
Cell Cycle ; 12(6): 872-6, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23439428

RESUMEN

Human sirtuin1 (SIRT1), the closest homolog of the yeast sir2 protein, functions as an NAD+-dependent histone and non-histone protein deacetylase in several cellular processes, like energy metabolism, stress responses, aging, etc. In our recent study, we have shown that lamin A (a major nuclear matrix protein) directly binds with and activates SIRT1. Resveratrol, a natural phenol, has long been known as an activator of SIRT1. However, resveratrol's direct activation of SIRT1 has been refuted several times. In our study, we have provided a mechanistic explanation to this question, and have shown that resveratrol activates SIRT1 by increasing its binding with lamin A, thus aiding in the nuclear matrix (NM) localization of SIRT1. We have also shown that rescue of adult stem cell (ASC) decline in laminopathy-based premature aging mice by resveratrol is SIRT1-dependent. Further, resveratrol's ameliorating effects on progeria and its capacity to extend lifespan in progeria mice has been established. Here we have summarized these findings and their probable implications on other aspects, like chromatin remodeling, stem cell therapy, DNA damage responses, etc.


Asunto(s)
Envejecimiento Prematuro/metabolismo , Lamina Tipo A/metabolismo , Progeria/metabolismo , Sirtuina 1/metabolismo , Estilbenos/farmacología , Células Madre Adultas/metabolismo , Envejecimiento Prematuro/tratamiento farmacológico , Animales , Línea Celular , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Histona Desacetilasas/metabolismo , Humanos , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Ratones , Ratones Noqueados , Matriz Nuclear/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progeria/tratamiento farmacológico , Resveratrol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA