Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Adv ; 10(20): eado1463, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758782

RESUMEN

A ketogenic diet (KD) is a high-fat, low-carbohydrate diet that leads to the generation of ketones. While KDs improve certain health conditions and are popular for weight loss, detrimental effects have also been reported. Here, we show mice on two different KDs and, at different ages, induce cellular senescence in multiple organs, including the heart and kidney. This effect is mediated through adenosine monophosphate-activated protein kinase (AMPK) and inactivation of mouse double minute 2 (MDM2) by caspase-2, leading to p53 accumulation and p21 induction. This was established using p53 and caspase-2 knockout mice and inhibitors to AMPK, p21, and caspase-2. In addition, senescence-associated secretory phenotype biomarkers were elevated in serum from mice on a KD and in plasma samples from patients on a KD clinical trial. Cellular senescence was eliminated by a senolytic and prevented by an intermittent KD. These results have important clinical implications, suggesting that the effects of a KD are contextual and likely require individual optimization.


Asunto(s)
Senescencia Celular , Dieta Cetogénica , Ratones Noqueados , Proteína p53 Supresora de Tumor , Animales , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Ratones , Humanos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Masculino , Especificidad de Órganos
2.
Sci Adv ; 10(20): eadj5942, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38758779

RESUMEN

Acetyl-CoA synthetase short-chain family member 1 (ACSS1) uses acetate to generate mitochondrial acetyl-CoA and is regulated by deacetylation by sirtuin 3. We generated an ACSS1-acetylation (Ac) mimic mouse, where lysine-635 was mutated to glutamine (K635Q). Male Acss1K635Q/K635Q mice were smaller with higher metabolic rate and blood acetate and decreased liver/serum ATP and lactate levels. After a 48-hour fast, Acss1K635Q/K635Q mice presented hypothermia and liver aberrations, including enlargement, discoloration, lipid droplet accumulation, and microsteatosis, consistent with nonalcoholic fatty liver disease (NAFLD). RNA sequencing analysis suggested dysregulation of fatty acid metabolism, cellular senescence, and hepatic steatosis networks, consistent with NAFLD. Fasted Acss1K635Q/K635Q mouse livers showed increased fatty acid synthase (FASN) and stearoyl-CoA desaturase 1 (SCD1), both associated with NAFLD, and increased carbohydrate response element-binding protein binding to Fasn and Scd1 enhancer regions. Last, liver lipidomics showed elevated ceramide, lysophosphatidylethanolamine, and lysophosphatidylcholine, all associated with NAFLD. Thus, we propose that ACSS1-K635-Ac dysregulation leads to aberrant lipid metabolism, cellular senescence, and NAFLD.


Asunto(s)
Senescencia Celular , Mitocondrias , Enfermedad del Hígado Graso no Alcohólico , Estearoil-CoA Desaturasa , Animales , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Ratones , Senescencia Celular/genética , Acetilación , Mitocondrias/metabolismo , Estearoil-CoA Desaturasa/metabolismo , Estearoil-CoA Desaturasa/genética , Masculino , Acetato CoA Ligasa/metabolismo , Acetato CoA Ligasa/genética , Técnicas de Sustitución del Gen , Hígado/metabolismo , Hígado/patología , Metabolismo de los Lípidos , Sirtuina 3/metabolismo , Sirtuina 3/genética , Modelos Animales de Enfermedad , Coenzima A Ligasas , Acido Graso Sintasa Tipo I
3.
Nat Commun ; 14(1): 5021, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596266

RESUMEN

Protein translation (PT) declines with age in invertebrates, rodents, and humans. It has been assumed that elevated PT at young ages is beneficial to health and PT ends up dropping as a passive byproduct of aging. In Drosophila, we show that a transient elevation in PT during early-adulthood exerts long-lasting negative impacts on aging trajectories and proteostasis in later-life. Blocking the early-life PT elevation robustly improves life-/health-span and prevents age-related protein aggregation, whereas transiently inducing an early-life PT surge in long-lived fly strains abolishes their longevity/proteostasis benefits. The early-life PT elevation triggers proteostatic dysfunction, silences stress responses, and drives age-related functional decline via juvenile hormone-lipid transfer protein axis and germline signaling. Our findings suggest that PT is adaptively suppressed after early-adulthood, alleviating later-life proteostatic burden, slowing down age-related functional decline, and improving lifespan. Our work provides a theoretical framework for understanding how lifetime PT dynamics shape future aging trajectories.


Asunto(s)
Envejecimiento , Longevidad , Humanos , Animales , Adulto , Drosophila , Células Germinativas , Hormonas Juveniles , Biosíntesis de Proteínas
4.
Sci Adv ; 8(23): eabk2252, 2022 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-35675410

RESUMEN

The proteasome has key roles in neuronal proteostasis, including the removal of misfolded and oxidized proteins, presynaptic protein turnover, and synaptic efficacy and plasticity. Proteasome dysfunction is a prominent feature of Alzheimer's disease (AD). We show that prevention of proteasome dysfunction by genetic manipulation delays mortality, cell death, and cognitive deficits in fly and cell culture AD models. We developed a transgenic mouse with neuronal-specific proteasome overexpression that, when crossed with an AD mouse model, showed reduced mortality and cognitive deficits. To establish translational relevance, we developed a set of TAT-based proteasome-activating peptidomimetics that stably penetrated the blood-brain barrier and enhanced 20S/26S proteasome activity. These agonists protected against cell death, cognitive decline, and mortality in cell culture, fly, and mouse AD models. The protective effects of proteasome overexpression appear to be driven, at least in part, by the proteasome's increased turnover of the amyloid precursor protein along with the prevention of overall proteostatic dysfunction.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Drosophila melanogaster , Ratones , Ratones Transgénicos , Complejo de la Endopetidasa Proteasomal/metabolismo
5.
Aging Cell ; 18(5): e13005, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31334599

RESUMEN

Cognitive function declines with age throughout the animal kingdom, and increasing evidence shows that disruption of the proteasome system contributes to this deterioration. The proteasome has important roles in multiple aspects of the nervous system, including synapse function and plasticity, as well as preventing cell death and senescence. Previous studies have shown neuronal proteasome depletion and inhibition can result in neurodegeneration and cognitive deficits, but it is unclear if this pathway is a driver of neurodegeneration and cognitive decline in aging. We report that overexpression of the proteasome ß5 subunit enhances proteasome assembly and function. Significantly, we go on to show that neuronal-specific proteasome augmentation slows age-related declines in measures of learning, memory, and circadian rhythmicity. Surprisingly, neuronal-specific augmentation of proteasome function also produces a robust increase of lifespan in Drosophila melanogaster. Our findings appear specific to the nervous system; ubiquitous proteasome overexpression increases oxidative stress resistance but does not impact lifespan and is detrimental to some healthspan measures. These findings demonstrate a key role of the proteasome system in brain aging.


Asunto(s)
Envejecimiento/metabolismo , Disfunción Cognitiva/prevención & control , Drosophila melanogaster/enzimología , Drosophila melanogaster/fisiología , Longevidad , Neuronas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Disfunción Cognitiva/enzimología , Drosophila melanogaster/citología
6.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 285-297, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30419337

RESUMEN

The mitochondrial genome (mtDNA) represents a tiny fraction of the whole genome, comprising just 16.6 kilobases encoding 37 genes involved in oxidative phosphorylation and the mitochondrial translation machinery. Despite its small size, much interest has developed in recent years regarding the role of mtDNA as a determinant of both aging and age-associated diseases. A number of studies have presented compelling evidence for key roles of mtDNA in age-related pathology, although many are correlative rather than demonstrating cause. In this review we will evaluate the evidence supporting and opposing a role for mtDNA in age-associated functional declines and diseases. We provide an overview of mtDNA biology, damage and repair as well as the influence of mitochondrial haplogroups, epigenetics and maternal inheritance in aging and longevity.


Asunto(s)
Envejecimiento/genética , ADN Mitocondrial/genética , Enfermedad/genética , Animales , Daño del ADN , ADN Mitocondrial/química , Radicales Libres/metabolismo , Humanos , Patrón de Herencia/genética
7.
PLoS Genet ; 12(7): e1006133, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27420916

RESUMEN

Mitochondrial dysfunction underlies numerous age-related pathologies. In an effort to uncover how the detrimental effects of mitochondrial dysfunction might be alleviated, we examined how the nematode C. elegans not only adapts to disruption of the mitochondrial electron transport chain, but in many instances responds with extended lifespan. Studies have shown various retrograde responses are activated in these animals, including the well-studied ATFS-1-dependent mitochondrial unfolded protein response (UPRmt). Such processes fall under the greater rubric of cellular surveillance mechanisms. Here we identify a novel p38 signaling cascade that is required to extend life when the mitochondrial electron transport chain is disrupted in worms, and which is blocked by disruption of the Mitochondrial-associated Degradation (MAD) pathway. This novel cascade is defined by DLK-1 (MAP3K), SEK-3 (MAP2K), PMK-3 (MAPK) and the reporter gene Ptbb-6::GFP. Inhibition of known mitochondrial retrograde responses does not alter induction of Ptbb-6::GFP, instead induction of this reporter often occurs in counterpoint to activation of SKN-1, which we show is under the control of ATFS-1. In those mitochondrial bioenergetic mutants which activate Ptbb-6::GFP, we find that dlk-1, sek-3 and pmk-3 are all required for their life extension.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/fisiología , Regulación de la Expresión Génica , Quinasas Quinasa Quinasa PAM/fisiología , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por Mitógenos/fisiología , Animales , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/fisiología , Genes Reporteros , Proteínas Fluorescentes Verdes/metabolismo , Mutación , Interferencia de ARN , Transducción de Señal , Respuesta de Proteína Desplegada , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Aging Cell ; 15(2): 336-48, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26729005

RESUMEN

Disruption of mitochondrial respiration in the nematode Caenorhabditis elegans can extend lifespan. We previously showed that long-lived respiratory mutants generate elevated amounts of α-ketoacids. These compounds are structurally related to α-ketoglutarate, suggesting they may be biologically relevant. Here, we show that provision of several such metabolites to wild-type worms is sufficient to extend their life. At least one mode of action is through stabilization of hypoxia-inducible factor-1 (HIF-1). We also find that an α-ketoglutarate mimetic, 2,4-pyridinedicarboxylic acid (2,4-PDA), is alone sufficient to increase the lifespan of wild-type worms and this effect is blocked by removal of HIF-1. HIF-1 is constitutively active in isp-1(qm150) Mit mutants, and accordingly, 2,4-PDA does not further increase their lifespan. Incubation of mouse 3T3-L1 fibroblasts with life-prolonging α-ketoacids also results in HIF-1α stabilization. We propose that metabolites that build up following mitochondrial respiratory dysfunction form a novel mode of cell signaling that acts to regulate lifespan.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácidos Cetoglutáricos/metabolismo , Longevidad/fisiología , Mitocondrias/metabolismo , Células 3T3-L1 , Animales , Ratones
9.
Brain Res ; 1621: 38-50, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-25452022

RESUMEN

This review covers the spatial and temporal rules governing induction of hippocampal long-term potentiation (LTP) by theta-burst stimulation. Induction of LTP in field CA1 by high frequency stimulation bursts that resemble the burst discharges (complex-spikes) of hippocampal pyramidal neurons involves a multiple-step mechanism. A single burst is insufficient for LTP induction because it evokes both excitatory and inhibitory currents that partially cancel and limit postsynaptic depolarization. Bursts repeated at the frequency (~5 Hz) of the endogenous theta rhythm induce maximal LTP, primarily because this frequency disables feed-forward inhibition and allows sufficient postsynaptic depolarization to activate voltage-sensitive NMDA receptors. The disinhibitory process, referred to as "priming", involves presynaptic GABA autoreceptors that inhibit GABA release. Activation of NMDA receptors allows a calcium flux into dendritic spines that serves as the proximal trigger for LTP. We include new data showing that theta-burst stimulation is more efficient than other forms of stimulation for LTP induction. In addition, we demonstrate that associative interactions between synapses activated during theta-bursts are limited to major dendritic domains since such interactions occur within apical or basal dendritic trees but not between them. We review evidence that recordings of electrophysiological responses during theta burst stimulation can help to determine if experimental manipulations that affect LTP do so by affecting events antecedent to the induction process, such as NMDA receptor activation, or downstream signaling cascades that result from postsynaptic calcium fluxes. Finally, we argue that theta-burst LTP represents a minimal model for stable, non-decremental LTP that is more sensitive to a variety of experimental manipulations than is LTP induced by other stimulation paradigms. This article is part of a Special Issue entitled SI: Brain and Memory.


Asunto(s)
Estimulación Eléctrica/métodos , Hipocampo/fisiología , Potenciación a Largo Plazo , Animales , Humanos , Potenciales de la Membrana , Células Piramidales/fisiología , Sinapsis/fisiología
10.
Exp Gerontol ; 56: 221-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24699406

RESUMEN

Mitochondria play numerous, essential roles in the life of eukaryotes. Disruption of mitochondrial function in humans is often pathological or even lethal. Surprisingly, in some organisms mitochondrial dysfunction can result in life extension. This paradox has been studied most extensively in the long-lived Mit mutants of the nematode Caenorhabditis elegans. In this review, we explore the major responses that are activated following mitochondrial dysfunction in these animals and how these responses potentially act to extend their life. We focus our attention on five broad areas of current research--reactive oxygen species signaling, the mitochondrial unfolded protein response, autophagy, metabolic adaptation, and the roles played by various transcription factors. Lastly, we also examine why disruption of complexes I and II differ in their ability to induce the Mit phenotype and extend lifespan.


Asunto(s)
Envejecimiento/metabolismo , Metabolismo Energético , Longevidad , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Factores de Edad , Envejecimiento/genética , Envejecimiento/patología , Animales , Autofagia , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Metabolismo Energético/genética , Genotipo , Humanos , Mitocondrias/patología , Proteínas Mitocondriales/genética , Modelos Animales , Estrés Oxidativo , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada
11.
Aging (Albany NY) ; 5(10): 741-58, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24107417

RESUMEN

While numerous life-extending manipulations have been discovered in the nematode Caenorhabditis elegans, one that remains most enigmatic is disruption of oxidative phosphorylation. In order to unravel how such an ostensibly deleterious manipulation can extend lifespan, we sought to identify the ensemble of nuclear transcription factors that are activated in response to defective mitochondrial electron transport chain (ETC) function. Using a feeding RNAi approach, we targeted over 400 transcription factors and identified 15 that, when reduced in function, reproducibly and differentially altered the development, stress response, and/or fecundity of isp-1(qm150) Mit mutants relative to wild-type animals. Seven of these transcription factors--AHA-1, CEH-18, HIF-1, JUN-1, NHR-27, NHR-49 and the CREB homolog-1 (CRH-1)-interacting protein TAF-4--were also essential for isp-1 life extension. When we tested the involvement of these seven transcription factors in the life extension of two other Mit mutants, namely clk-1(qm30) and tpk-1(qm162), TAF-4 and HIF-1 were consistently required. Our findings suggest that the Mit phenotype is under the control of multiple transcriptional responses, and that TAF-4 and HIF-1 may be part of a general signaling axis that specifies Mit mutant life extension.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Complejo III de Transporte de Electrones/fisiología , Longevidad , Mitocondrias/metabolismo , Tiamina Pirofosfoquinasa/fisiología , Factores de Transcripción/fisiología , Animales , Caenorhabditis elegans/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Transporte de Electrón , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...