Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 313
Filtrar
1.
Cell ; 165(6): 1361-1374, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259148

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare, invariably fatal premature aging disorder. The disease is caused by constitutive production of progerin, a mutant form of the nuclear architectural protein lamin A, leading, through unknown mechanisms, to diverse morphological, epigenetic, and genomic damage and to mesenchymal stem cell (MSC) attrition in vivo. Using a high-throughput siRNA screen, we identify the NRF2 antioxidant pathway as a driver mechanism in HGPS. Progerin sequesters NRF2 and thereby causes its subnuclear mislocalization, resulting in impaired NRF2 transcriptional activity and consequently increased chronic oxidative stress. Suppressed NRF2 activity or increased oxidative stress is sufficient to recapitulate HGPS aging defects, whereas reactivation of NRF2 activity in HGPS patient cells reverses progerin-associated nuclear aging defects and restores in vivo viability of MSCs in an animal model. These findings identify repression of the NRF2-mediated antioxidative response as a key contributor to the premature aging phenotype.


Asunto(s)
Envejecimiento Prematuro/metabolismo , Antioxidantes/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Progeria/metabolismo , Envejecimiento Prematuro/genética , Línea Celular , Supervivencia Celular , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Factor 2 Relacionado con NF-E2/genética , Progeria/genética , ARN Interferente Pequeño , Factores de Transcripción/metabolismo , Transcripción Genética
2.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27984723

RESUMEN

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Asunto(s)
Envejecimiento/genética , Reprogramación Celular , Epigénesis Genética , Enfermedades Metabólicas/genética , Factores de Transcripción/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Animales , Diabetes Mellitus Tipo 2/inducido químicamente , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Lamina Tipo A/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/prevención & control , Ratones , Modelos Animales , Páncreas/metabolismo , Sarcopenia/metabolismo
3.
Nat Rev Mol Cell Biol ; 18(10): 595-609, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28792007

RESUMEN

Ageing is the predominant risk factor for many common diseases. Human premature ageing diseases are powerful model systems to identify and characterize cellular mechanisms that underpin physiological ageing. Their study also leads to a better understanding of the causes, drivers and potential therapeutic strategies of common diseases associated with ageing, including neurological disorders, diabetes, cardiovascular diseases and cancer. Using the rare premature ageing disorder Hutchinson-Gilford progeria syndrome as a paradigm, we discuss here the shared mechanisms between premature ageing and ageing-associated diseases, including defects in genetic, epigenetic and metabolic pathways; mitochondrial and protein homeostasis; cell cycle; and stem cell-regenerative capacity.


Asunto(s)
Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento/genética , Envejecimiento Prematuro/genética , Animales , Reparación del ADN , Epigénesis Genética , Inestabilidad Genómica , Humanos , Progeria/genética , Progeria/metabolismo , Progeria/patología
4.
Stem Cells ; 41(1): 39-49, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36219686

RESUMEN

Mitochondria are indispensable in maintaining hematopoietic stem cells (HSCs), and mitochondrial complex II (MCII) has been recognized as a key component of HSCs. However, the physiological role of MCII on long-term hematopoiesis and hematopoietic reconstitution capacity remains unknown. Hence, this study evaluated the impact of MCII dysfunctions on long-term HSC maintenance and hematopoietic homeostasis among conditional transgenic mice with a missense mutation in the succinate dehydrogenase complex subunit C gene (SdhcV69E). HSCs collected from SdhcV69E mice had a higher reactive oxygen species (ROS) accumulation and DNA damage in response to mitochondrial activation. Via the aging stress response, MCII dysfunctions caused decreased white blood cell count with myeloid-skewing property, macrocytic anemia, and thrombocytosis. Moreover, the HSCs of aged SdhcV69E mice exhibited greater ROS accumulation and lower membrane potential. Transplantation-induced replicative stress also caused premature senescent hematopoiesis. Furthermore, accelerated ROS accumulation and profound DNA damage in HSCs were observed in the SdhcV69E-derived cell recipients. The long-term hematopoietic reconstitution capacity was remarkably impaired in HSCs from the SdhcV69E-derived cell recipients. Taken together, MCII plays an essential role in long-term hematopoiesis, and MCII dysfunctions with aging or replicative stresses caused excessive ROS accumulation and DNA damage in HSCs, leading to premature senescence.


Asunto(s)
Envejecimiento Prematuro , Ratones , Animales , Especies Reactivas de Oxígeno/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Transporte de Electrón , Células Madre Hematopoyéticas/metabolismo , Envejecimiento/genética , Ratones Transgénicos , Hematopoyesis/genética , Ratones Endogámicos C57BL
5.
FASEB J ; 37(8): e23116, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37498235

RESUMEN

Laminopathies are a group of rare genetic disorders with heterogeneous clinical phenotypes such as premature aging, cardiomyopathy, lipodystrophy, muscular dystrophy, microcephaly, epilepsy, and so on. The cellular phenomena associated with laminopathy invariably show disruption of nucleoskeleton of lamina due to deregulated expression, localization, function, and interaction of mutant lamin proteins. Impaired spatial and temporal tethering of lamin proteins to the lamina or nucleoplasmic aggregation of lamins are the primary molecular events that can trigger nuclear proteotoxicity by modulating differential protein-protein interactions, sequestering quality control proteins, and initiating a cascade of abnormal post-translational modifications. Clearly, laminopathic cells exhibit moderate to high nuclear proteotoxicity, raising the question of whether an imbalance in nuclear proteostasis is involved in laminopathic diseases, particularly in diseases of early aging such as HGPS and laminopathy-associated premature aging. Here, we review nuclear proteostasis and its deregulation in the context of lamin proteins and laminopathies.


Asunto(s)
Envejecimiento Prematuro , Laminopatías , Humanos , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Proteostasis , Núcleo Celular/metabolismo , Laminas/genética , Laminas/metabolismo , Laminopatías/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Mutación , Lámina Nuclear/genética , Lámina Nuclear/metabolismo
6.
Nucleic Acids Res ; 50(16): 9260-9278, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36039758

RESUMEN

Nestor-Guillermo progeria syndrome (NGPS) is caused by a homozygous alanine-to-threonine mutation at position 12 (A12T) in barrier-to-autointegration factor (BAF). It is characterized by accelerated aging with severe skeletal abnormalities. BAF is an essential protein binding to DNA and nuclear envelope (NE) proteins, involved in NE rupture repair. Here, we assessed the impact of BAF A12T on NE integrity using NGPS-derived patient fibroblasts. We observed a strong defect in lamin A/C accumulation to NE ruptures in NGPS cells, restored upon homozygous reversion of the pathogenic BAF A12T mutation with CRISPR/Cas9. By combining in vitro and cellular assays, we demonstrated that while the A12T mutation does not affect BAF 3D structure and phosphorylation by VRK1, it specifically decreases the interaction between BAF and lamin A/C. Finally, we revealed that the disrupted interaction does not prevent repair of NE ruptures but instead generates weak points in the NE that lead to a higher frequency of NE re-rupturing in NGPS cells. We propose that this NE fragility could directly contribute to the premature aging phenotype in patients.


Asunto(s)
Envejecimiento Prematuro , Progeria , Humanos , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Progeria/metabolismo , Envejecimiento Prematuro/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Unión al ADN/genética , Mutación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas , Péptidos y Proteínas de Señalización Intracelular/metabolismo
7.
Int J Mol Sci ; 25(3)2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38338968

RESUMEN

The primary neuronal and astrocyte culture described here is from the stress-hyperreactive Wistar Kyoto (WKY) More Immobile (WMI) rat with premature aging-related memory deficit, and its nearly isogenic control, the Less Immobile (WLI) strain. Primary WMI hippocampal neurons and cortical astrocytes are significantly more sensitive to oxidative stress (OS) generated by administration of H2O2 compared to WLI cells as measured by the trypan blue cell viability assay. Intrinsic genetic vulnerability is also suggested by the decreased gene expression in WMI neurons of catalase (Cat), and in WMI cortical astrocytes of insulin-like growth factor 2 (Igf2), synuclein gamma (Sncg) and glutathione peroxidase 2 (Gpx2) compared to WLI. The expressions of several mitochondrial genes are dramatically increased in response to H2O2 treatment in WLI, but not in WMI cortical astrocytes. We propose that the vulnerability of WMI neurons to OS is due to the genetic differences between the WLI and WMI. Furthermore, the upregulation of mitochondrial genes may be a compensatory response to the generation of free radicals by OS in the WLIs, and this mechanism is disturbed in the WMIs. Thus, this pilot study suggests intrinsic vulnerabilities in the WMI hippocampal neurons and cortical astrocytes, and affirm the efficacy of this bimodal in vitro screening system for finding novel drug targets to prevent oxidative damage in illnesses.


Asunto(s)
Envejecimiento Prematuro , Envejecimiento Cognitivo , Ratas , Animales , Ratas Endogámicas WKY , Astrocitos/metabolismo , Envejecimiento Prematuro/metabolismo , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/metabolismo , Proyectos Piloto , Estrés Oxidativo , Neuronas/metabolismo , Células Cultivadas
8.
Plant Cell Environ ; 46(3): 1004-1017, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36515398

RESUMEN

Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in immune response in animals. However, the role of MIFs in plants such as Medicago truncatula, particularly in symbiotic nitrogen fixation, remains unclear. An investigation of M. truncatula-Sinorhizobium meliloti symbiosis revealed that MtMIF3 was mainly expressed in the nitrogen-fixing zone of the nodules. Silencing MtMIF3 using RNA interference (Ri) technology resulted in increased nodule numbers but higher levels of bacteroid degradation in the infected cells of the nitrogen-fixing zone, suggesting that premature aging was induced in MtMIF3-Ri nodules. In agreement with this conclusion, the activities of nitrogenase, superoxide dismutase and catalase were lower than those in controls, but cysteine proteinase activity was increased in nodulated roots at 28 days postinoculation. In contrast, the overexpression of MtMIF3 inhibited nodule senescence. MtMIF3 is localized in the plasma membrane, nucleus, and cytoplasm, where it interacts with methionine sulfoxide reductase B (MsrB), which is also localized in the chloroplasts of tobacco leaf cells. Taken together, these results suggest that MtMIF3 prevents premature nodule aging and protects against oxidation by interacting with MtMsrB.


Asunto(s)
Envejecimiento Prematuro , Factores Inhibidores de la Migración de Macrófagos , Medicago truncatula , Nódulos de las Raíces de las Plantas/metabolismo , Medicago truncatula/fisiología , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Envejecimiento Prematuro/metabolismo , Fijación del Nitrógeno/fisiología , Nitrógeno/metabolismo , Simbiosis/fisiología
9.
Int J Mol Sci ; 24(5)2023 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-36902379

RESUMEN

Digital stress is a newly identified cosmetic stress that is mainly characterized by blue light exposure. The effects of this stress have become increasingly important with the emergence of personal digital devices, and its deleterious effects on the body are now well-known. Blue light has been observed to cause perturbation of the natural melatonin cycle and skin damage similar to that from UVA exposure, thus leading to premature aging. "A melatonin-like ingredient" was discovered in the extract of Gardenia jasminoides, which acts as a filter against blue light and as a melatonin-like ingredient to prevent and stop premature aging. The extract showed significant protective effects on the mitochondrial network of primary fibroblasts, a significant decrease of -86% in oxidized proteins on skin explants, and preservation of the natural melatonin cycle in the co-cultures of sensory neurons and keratinocytes. Upon analysis using in silico methods, only the crocetin form, released through skin microbiota activation, was found to act as a melatonin-like molecule by interacting with the MT1-receptor, thus confirming its melatonin-like properties. Finally, clinical studies revealed a significant decrease in wrinkle number of -21% in comparison to the placebo. The extract showed strong protection against blue light damage and the prevention of premature aging through its melatonin-like properties.


Asunto(s)
Envejecimiento Prematuro , Gardenia , Melatonina , Envejecimiento Prematuro/metabolismo , Melatonina/farmacología , Piel/metabolismo
10.
Int J Mol Sci ; 24(3)2023 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-36768831

RESUMEN

Chronic Kidney Disease (CKD), a global health burden, is strongly associated with age-related renal function decline, hypertension, and diabetes, which are all frequent consequences of obesity. Despite extensive studies, the mechanisms determining susceptibility to CKD remain insufficiently understood. Clinical evidence together with prior studies from our group showed that perinatal metabolic disorders after intrauterine growth restriction or maternal obesity adversely affect kidney structure and function throughout life. Since obesity and aging processes converge in similar pathways we tested if perinatal obesity caused by high-fat diet (HFD)-fed dams sensitizes aging-associated mechanisms in kidneys of newborn mice. The results showed a marked increase of γH2AX-positive cells with elevated 8-Oxo-dG (RNA/DNA damage), both indicative of DNA damage response and oxidative stress. Using unbiased comprehensive transcriptomics we identified compartment-specific differentially-regulated signaling pathways in kidneys after perinatal obesity. Comparison of these data to transcriptomic data of naturally aged kidneys and prematurely aged kidneys of genetic modified mice with a hypomorphic allele of Ercc1, revealed similar signatures, e.g., inflammatory signaling. In a biochemical approach we validated pathways of inflammaging in the kidneys after perinatal obesity. Collectively, our initial findings demonstrate premature aging-associated processes as a consequence of perinatal obesity that could determine the susceptibility for CKD early in life.


Asunto(s)
Envejecimiento Prematuro , Insuficiencia Renal Crónica , Femenino , Ratones , Animales , Embarazo , Humanos , Envejecimiento Prematuro/metabolismo , Obesidad/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Dieta Alta en Grasa/efectos adversos , Envejecimiento/genética
11.
Int J Cosmet Sci ; 45(6): 791-801, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37602524

RESUMEN

OBJECTIVE: It is well established that exposure of human skin to airborne pollution, particularly in the form of particulate matter sized 2.5 µm (PM2.5 ), is associated with oxidative stress, DNA damage and inflammation, leading to premature signs of skin aging. Because much of the damage results from oxidative stress, we examined the effects of a topical composition containing three antioxidants in an in vitro model system to assess the potential for amelioration of premature aging. The use of multiple antioxidants was of interest based on the typical composition of therapeutic skincare products. It is important to determine the efficacy of multiple antioxidants together and develop a short-term assay for larger scale efficacy testing. METHODS: Normal human epidermal keratinocytes were exposed to a rural-derived source of PM2.5 in the presence and absence of an antioxidant mixture of resveratrol, niacinamide and GHK peptide. Endpoints related to inflammation, premature aging and carcinogenicity were monitored after 5 h of exposure and included IL-6, CXCL10, MMP-1 and NRF2. Differentially expressed genes were monitored by RNA-seq. RESULTS: Pre-treatment of keratinocytes with the antioxidant preparation in the absence of PM2.5 reduced baseline levels of MMP-1, IL-6 and CYP1A1 and reduced PM2.5 -induced increases in all four endpoints, MMP-1, IL-6, CXCL10 and CYP1A1. Antioxidants significantly increased NRF2 protein in the presence of PM2.5 , indicating a protective response. RNA-seq interrogation of antioxidant-treated cells further showed increased expression of NRF2 inducible genes. The expression of CYP1A1 and genes related to aryl hydrocarbon activation were induced by PM2.5 and suppressed by antioxidants. CONCLUSIONS: Specific signalling pathways known to be correlated with skin inflammation and aging were examined based on their suitability for use in efficacy testing for the prevention of skin damage due to ambient hydrocarbon pollution. Endpoints examined after only 5 h of exposure provide a useful method amenable to high through-put screening. The results obtained reinforce the concept that a multiple antioxidant preparation, topically applied, may reduce pro-inflammatory signalling and cellular damage and thereby reduce premature skin aging due to exposure to rural-derived airborne pollution.


OBJECTIF: Il est bien établi que l'exposition de la peau humaine à la pollution atmosphérique, en particulier sous forme de particules d'une taille de 2,5 µm (PM2,5 ), est associée à un stress oxydatif, à des dommages à l'ADN et à une inflammation entraînant des signes prématurés de vieillissement cutané. Étant donné que la plupart des dommages résultent du stress oxydatif, nous avons examiné les effets d'une composition topique contenant trois antioxydants dans un système de modèle in vitro afin d'évaluer le potentiel d'amélioration du vieillissement prématuré. L'utilisation de plusieurs antioxydants a été intéressante en raison de la composition typique des produits thérapeutiques de soin de la peau. Il est important de déterminer l'efficacité de plusieurs antioxydants combinés et de développer un test à court terme pour des tests d'efficacité à plus grande échelle. MÉTHODES: Des kératinocytes épidermiques humains normaux ont été exposés à une source de PM2,5 rurale en présence et en l'absence d'un mélange antioxydant de resvératrol, de niacinamide et de peptide GHK. Les critères d'évaluation liés à l'inflammation, au vieillissement prématuré et à la carcinogénicité ont été surveillés après 5 heures d'exposition et comprenaient l'IL-6, CXCL10, MMP-1 et le NRF2. Les gènes exprimés de manière différentielle ont été surveillés par séquençage de l'ARN. RÉSULTATS: Le prétraitement des kératinocytes par la préparation antioxydante en l'absence de PM2,5 a réduit les taux initiaux de MMP-1, IL-6 et de CYP1A1 et a réduit les augmentations induites par les PM2,5 dans les quatre critères d'évaluation, MMP-1, IL-6, CXCL10 et CYP1A1. Les antioxydants ont significativement augmenté la protéine NRF2 en présence de PM2,5 , ce qui indique une réponse protectrice. L'interrogation des séquences d'ARN des cellules traitées par antioxydants a également montré une expression accrue des gènes inductibles par NRF2. L'expression du CYP1A1 et des gènes liés à l'activation des hydrocarbures aryles a été induite par les PM2,5 et supprimée par les antioxydants. CONCLUSIONS: Les voies de signalisation spécifiques connues pour être corrélées à l'inflammation cutanée et au vieillissement ont été examinées en fonction de leur adéquation à l'utilisation dans les tests d'efficacité pour la prévention des lésions cutanées dues à la pollution des hydrocarbures ambiants. Les critères d'évaluation examinés après seulement 5 heures d'exposition fournissent une méthode utile pouvant être utilisée pour un dépistage à haut débit. Les résultats obtenus renforcent le principe selon lequel une préparation antioxydante multiple, appliquée par voie topique, peut réduire la signalisation pro-inflammatoire et les dommages cellulaires et ainsi réduire le vieillissement prématuré de la peau résultant de l'exposition à la pollution atmosphérique d'origine rurale.


Asunto(s)
Envejecimiento Prematuro , Antioxidantes , Humanos , Antioxidantes/farmacología , Antioxidantes/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Envejecimiento Prematuro/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/farmacología , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A1/farmacología , Interleucina-6/metabolismo , Interleucina-6/farmacología , Queratinocitos , Material Particulado/toxicidad , Estrés Oxidativo , Resveratrol/farmacología , Polvo , Inflamación
12.
Blood ; 135(23): 2049-2058, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32305044

RESUMEN

Loss of heterochromatin has been proposed as a universal mechanism of aging across different species and cell types. However, a comprehensive analysis of hematopoietic changes caused by heterochromatin loss is lacking. Moreover, there is conflict in the literature around the role of the major heterochromatic histone methyltransferase Suv39h1 in the aging process. Here, we use individual and dual deletion of Suv39h1 and Suv39h2 enzymes to examine the causal role of heterochromatin loss in hematopoietic cell development. Loss of neither Suv39h1 nor Suv39h2 individually had any effect on hematopoietic stem cell function or the development of mature lymphoid or myeloid lineages. However, deletion of both enzymes resulted in characteristic changes associated with aging such as reduced hematopoietic stem cell function, thymic involution and decreased lymphoid output with a skewing toward myeloid development, and increased memory T cells at the expense of naive T cells. These cellular changes were accompanied by molecular changes consistent with aging, including alterations in nuclear shape and increased nucleolar size. Together, our results indicate that the hematopoietic system has a remarkable tolerance for major disruptions in chromatin structure and reveal a role for Suv39h2 in depositing sufficient H3K9me3 to protect the entire hematopoietic system from changes associated with premature aging.


Asunto(s)
Envejecimiento Prematuro/patología , Diferenciación Celular , Hematopoyesis , Células Madre Hematopoyéticas/patología , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Metiltransferasas/fisiología , Proteínas Represoras/fisiología , Anciano , Envejecimiento Prematuro/metabolismo , Animales , Núcleo Celular/genética , Femenino , Células Madre Hematopoyéticas/metabolismo , Heterocromatina/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
13.
PLoS Biol ; 17(10): e3000508, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31593566

RESUMEN

CDGSH iron-sulfur domain-containing protein 2 (Cisd2) is pivotal to mitochondrial integrity and intracellular Ca2+ homeostasis. In the heart of Cisd2 knockout mice, Cisd2 deficiency causes intercalated disc defects and leads to degeneration of the mitochondria and sarcomeres, thereby impairing its electromechanical functioning. Furthermore, Cisd2 deficiency disrupts Ca2+ homeostasis via dysregulation of sarco/endoplasmic reticulum Ca2+-ATPase (Serca2a) activity, resulting in an increased level of basal cytosolic Ca2+ and mitochondrial Ca2+ overload in cardiomyocytes. Most strikingly, in Cisd2 transgenic mice, a persistently high level of Cisd2 is sufficient to delay cardiac aging and attenuate age-related structural defects and functional decline. In addition, it results in a younger cardiac transcriptome pattern during old age. Our findings indicate that Cisd2 plays an essential role in cardiac aging and in the heart's electromechanical functioning. They highlight Cisd2 as a novel drug target when developing therapies to delay cardiac aging and ameliorate age-related cardiac dysfunction.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento/fisiología , Bloqueo Atrioventricular/genética , Proteínas Relacionadas con la Autofagia/genética , Corazón/fisiopatología , Proteínas del Tejido Nervioso/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/fisiopatología , Animales , Bloqueo Atrioventricular/diagnóstico por imagen , Bloqueo Atrioventricular/metabolismo , Bloqueo Atrioventricular/fisiopatología , Proteínas Relacionadas con la Autofagia/deficiencia , Calcio/metabolismo , Electrocardiografía , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Corazón/fisiología , Homeostasis/fisiología , Masculino , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/deficiencia , Sarcómeros/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Transcriptoma
14.
J Assist Reprod Genet ; 39(9): 2143-2151, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35881273

RESUMEN

BACKGROUND: To explore the differences between a population with premature endometrial aging and a population with normal endometrial status in young women with recurrent implantation failure (< 35 years). METHODS: Systematic analysis of the endometrium transcriptome of 274 RIF women. The NMF algorithm was used for classification based on endometrial-specific aging markers in CellAge, and the endometrial receptivity, gene expression patterns, and clinical data were compared between the classifications. RESULTS: Two hundred forty-five young RIF women could be divided into two clusters, in which the aging gene expression pattern of cluster 2 was closer to the reference cluster. Cluster 1 was characterized by high immune activity, while cluster 2 was characterized by high metabolic activity. Combined with clinical data, cluster 2 was worse than cluster 1 in window of implantation deviation rate and endometrial receptivity. CONCLUSION: Premature aging of the endometrium exists in young women with RIF, and premature aging of the endometrium was associated with poor reproductive outcomes.


Asunto(s)
Envejecimiento Prematuro , Infertilidad Femenina , Envejecimiento/genética , Envejecimiento Prematuro/metabolismo , Biomarcadores/metabolismo , Implantación del Embrión/genética , Endometrio/metabolismo , Femenino , Humanos , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo
15.
Int J Mol Sci ; 23(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35269833

RESUMEN

Both intrinsic (i.e., an individual's body clock) and extrinsic factors (i.e., air pollutants and ultraviolet irradiation) accelerate premature aging. Epidemiological studies have shown a correlation between pollutant levels and aging skin symptoms. Diesel particle matter in particular leads to some diseases, including in the skin. Our recent study demonstrates that diesel particulate extract (DPE) increases apoptosis via increases in an anti-mitogenic/pro-apoptotic lipid mediator, ceramide in epidermal keratinocytes. Here, we investigated whether and how DPE accelerates premature skin aging using cultured normal human dermal fibroblasts (HDF). We first demonstrated that DPE increases cell senescence marker ß-galactosidase activity in HDF. We then found increases in mRNA and protein levels, along with activity of matrix metalloprotease (MMP)-1 and MMP-3, which are associated with skin aging following DPE exposure. We confirmed increases in collagen degradation in HDF treated with DPE. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) is activated by DPE and results in increased ceramide production by sphingomyelinase activation in HDF. We identified that ceramide-1-phosphate (C1P) (produced from ceramide by ceramide kinase activation) activates MMP-1 and MMP-3 through activation of arachidonate cascade, followed by STAT 1- and STAT 3-dependent transcriptional activation.


Asunto(s)
Envejecimiento Prematuro , Envejecimiento de la Piel , Envejecimiento Prematuro/metabolismo , Células Cultivadas , Ceramidas/metabolismo , Fibroblastos/metabolismo , Humanos , Metaloproteinasa 3 de la Matriz/metabolismo , NADPH Oxidasas/metabolismo , Fosfatos/metabolismo , Extractos Vegetales/metabolismo , Transducción de Señal , Piel/metabolismo , Rayos Ultravioleta/efectos adversos
16.
Exp Eye Res ; 209: 108698, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34228964

RESUMEN

Vision requires the transport and recycling of the pigment 11-cis retinaldehyde (retinal) between the retinal pigment epithelium (RPE) and photoreceptors. 11-cis retinal is also required for light-mediated photoreceptor death in dark-adapted mouse eye, probably through overstimulation of rod cells adapted for low light. Retbindin is a photoreceptor-specific protein, of unclear function, that is localized between the RPE and the tips of the photoreceptors. Unexpectedly, young Rtbdn-KO mice, with targeted deletion (KO) of retbindin, showed delayed regeneration of retinal function after bleaching and were strongly resistant to light-induced photoreceptor death. Furthermore, bio-layer interferometry binding studies showed recombinant retbindin had significant affinity for retinoids, most notably 11-cis retinal. This suggests that retbindin mediates light damage, probably through a role in transport of 11-cis retinal. In Rtbdn-KO mice, retinal development was normal, as were amplitudes of rod and cone electroretinograms (ERG) up to 4 months, although implicit times and c-waves were affected. However, with aging, both light- and dark-adapted ERG amplitudes declined significantly and photoreceptor outer segments became disordered, However, in contrast to other reports, there was little retinal degeneration or drop in flavin levels. The RPE developed vacuoles and lipid, protein and calcium deposits reminiscent of age-related macular degeneration. Other signs of premature aging included loss of OPN4+ retinal ganglion cells and activation of microglia. Thus, retbindin plays an unexpected role in the mammalian visual cycle, probably as an adaptation for vision in dim light. It mediates light damage in the dark-adapted eye, but also plays a role in light-adapted responses and in long term retinal homeostasis.


Asunto(s)
Envejecimiento Prematuro/genética , Proteínas del Ojo/genética , Regulación de la Expresión Génica , ARN/genética , Células Fotorreceptoras Retinianas Conos/metabolismo , Enfermedades de la Retina/genética , Epitelio Pigmentado de la Retina/metabolismo , Envejecimiento Prematuro/metabolismo , Animales , Adaptación a la Oscuridad/fisiología , Modelos Animales de Enfermedad , Electrorretinografía , Proteínas del Ojo/biosíntesis , Ratones , Microscopía Electrónica de Transmisión , Células Fotorreceptoras Retinianas Conos/ultraestructura , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Epitelio Pigmentado de la Retina/ultraestructura
17.
FASEB J ; 34(6): 7905-7914, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32282093

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) arises when a truncated form of farnesylated prelamin A accumulates at the nuclear envelope, leading to misshapen nuclei. Previous studies of adult Zmpste24-deficient mice, a mouse model of progeria, have reported a metabolic response involving inhibition of the mTOR (mammalian target of rapamycin) kinase and activation of autophagy. However, exactly how mTOR or autophagy is involved in progeria remains unclear. Here, we investigate this question by crossing Zmpste24+/- mice with mice hypomorphic in mTOR (mTOR△/+ ), or mice heterozygous in autophagy-related gene 7 (Atg7+/- ). We find that accumulation of prelamin A induces premature aging through mTOR overactivation and impaired autophagy in newborn Zmpste24-/- mice. Zmpste24-/- mice with genetically reduced mTOR activity, but not heterozygosity in Atg7, show extended lifespan. Moreover, mTOR inhibition partially restores autophagy and S6K1 activity. We also show that progerin interacts with the Akt phosphatase to promote full activation of the Akt/mTOR signaling pathway. Finally, although we find that genetic reduction of mTOR postpones premature aging in Zmpste24 KO mice, frequent embryonic lethality occurs. Together, our findings show that over-activated mTOR contributes to premature aging in Zmpste24-/- mice, and suggest a potential strategy in treating HGPS patients with mTOR inhibitors.


Asunto(s)
Envejecimiento Prematuro/metabolismo , Lamina Tipo A/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/fisiología , Proteína 7 Relacionada con la Autofagia/metabolismo , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Femenino , Fibroblastos/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Masculino , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Progeria/metabolismo , Transducción de Señal/fisiología
18.
Nucleic Acids Res ; 47(22): 11709-11728, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31647095

RESUMEN

The A-type lamins (lamin A/C), encoded by the LMNA gene, are important structural components of the nuclear lamina. LMNA mutations lead to degenerative disorders known as laminopathies, including the premature aging disease Hutchinson-Gilford progeria syndrome. In addition, altered lamin A/C expression is found in various cancers. Reports indicate that lamin A/C plays a role in DNA double strand break repair, but a role in DNA base excision repair (BER) has not been described. We provide evidence for reduced BER efficiency in lamin A/C-depleted cells (Lmna null MEFs and lamin A/C-knockdown U2OS). The mechanism involves impairment of the APE1 and POLß BER activities, partly effectuated by associated reduction in poly-ADP-ribose chain formation. Also, Lmna null MEFs displayed reduced expression of several core BER enzymes (PARP1, LIG3 and POLß). Absence of Lmna led to accumulation of 8-oxoguanine (8-oxoG) lesions, and to an increased frequency of substitution mutations induced by chronic oxidative stress including GC>TA transversions (a fingerprint of 8-oxoG:A mismatches). Collectively, our results provide novel insights into the functional interplay between the nuclear lamina and cellular defenses against oxidative DNA damage, with implications for cancer and aging.


Asunto(s)
Reparación del ADN/genética , Lamina Tipo A/fisiología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Animales , Células Cultivadas , Daño del ADN/fisiología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Análisis por Micromatrices , Lámina Nuclear/genética , Lámina Nuclear/metabolismo , Estrés Oxidativo/genética , Progeria/genética
19.
FASEB J ; 33(11): 12644-12654, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31483997

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor, largely known for its role in xenobiotic metabolism and detoxification as well as its crucial role as a regulator of inflammation. Here, we have compared a cohort wild-type and AhR-null mice along aging to study the relationship between this receptor and age-associated inflammation, termed as "inflammaging," both at a systemic and the CNS level. Our results show that AhR deficiency is associated with a premature aged phenotype, characterized by early inflammaging, as shown by an increase in plasma cytokines levels. The absence of AhR also promotes the appearance of brain aging anatomic features, such as the loss of the white matter integrity. In addition, AhR-/- mice present an earlier spatial memory impairment and an enhanced astrogliosis in the hippocampus when compared with their age-matched AhR+/+ controls. Importantly, we have found that AhR protein levels decrease with age in this brain structure, strongly suggesting a link between AhR and aging.-Bravo-Ferrer, I., Cuartero, M. I., Medina, V., Ahedo-Quero, D., Peña-Martínez, C., Pérez-Ruíz, A., Fernández-Valle, M. E., Hernández-Sánchez, C., Fernández-Salguero, P. M., Lizasoain, I., Moro, M. A. Lack of the aryl hydrocarbon receptor accelerates aging in mice.


Asunto(s)
Envejecimiento Prematuro , Envejecimiento , Hipocampo , Receptores de Hidrocarburo de Aril/deficiencia , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/patología , Animales , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratones , Ratones Noqueados , Receptores de Hidrocarburo de Aril/metabolismo , Memoria Espacial
20.
Int J Mol Sci ; 22(1)2020 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-33396477

RESUMEN

The vast majority of the literature on the aryl hydrocarbon receptor is concerned with its functions in xenobiotic detoxification. However, in the course of evolution, this receptor had to have physiological (rather than toxicological) functions. Our aim was to review the aryl hydrocarbon receptor's role in the physiological functions involved in aging. This study was performed by searching the MEDLINE and Google Academic databases. A total of 34 articles were selected that focused specifically on the aryl hydrocarbon receptor and aging, the aryl hydrocarbon receptor and physiological functions, and the combination of both. This receptor's main physiological functions (mediated by the modulation of gene expression) were cell regeneration, the immune reaction, intestinal homeostasis, and cell proliferation. Furthermore, it was shown that the loss of this receptor led to premature aging. This process may be caused by the dysregulation of hematopoietic stem cells, loss of glucose and lipid homeostasis, increase in inflammation, and deterioration of the brain. We conclude that the aryl hydrocarbon receptor, apart from its well-established role in xenobiotic detoxication, plays an important role in physiological functions and in the aging process. Modulation of the signaling pathway of this receptor could be a therapeutic target of interest in aging.


Asunto(s)
Envejecimiento Prematuro/prevención & control , Envejecimiento/patología , Receptores de Hidrocarburo de Aril/metabolismo , Xenobióticos/metabolismo , Envejecimiento Prematuro/metabolismo , Animales , Humanos , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA