Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nat Commun ; 15(1): 828, 2024 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-38280853

RESUMEN

Caloric Restriction (CR) has established anti-cancer effects, but its clinical relevance and molecular mechanism remain largely undefined. Here, we investigate CR's impact on several mouse models of Acute Myeloid Leukemias, including Acute Promyelocytic Leukemia, a subtype strongly affected by obesity. After an initial marked anti-tumor effect, lethal disease invariably re-emerges. Initially, CR leads to cell-cycle restriction, apoptosis, and inhibition of TOR and insulin/IGF1 signaling. The relapse, instead, is associated with the non-genetic selection of Leukemia Initiating Cells and the downregulation of double-stranded RNA (dsRNA) sensing and Interferon (IFN) signaling genes. The CR-induced adaptive phenotype is highly sensitive to pharmacological or genetic ablation of LSD1, a lysine demethylase regulating both stem cells and dsRNA/ IFN signaling. CR + LSD1 inhibition leads to the re-activation of dsRNA/IFN signaling, massive RNASEL-dependent apoptosis, and complete leukemia eradication in ~90% of mice. Importantly, CR-LSD1 interaction can be modeled in vivo and in vitro by combining LSD1 ablation with pharmacological inhibitors of insulin/IGF1 or dual PI3K/MEK blockade. Mechanistically, insulin/IGF1 inhibition sensitizes blasts to LSD1-induced death by inhibiting the anti-apoptotic factor CFLAR. CR and LSD1 inhibition also synergize in patient-derived AML and triple-negative breast cancer xenografts. Our data provide a rationale for epi-metabolic pharmacologic combinations across multiple tumors.


Asunto(s)
Insulinas , Leucemia Mieloide Aguda , Humanos , Animales , Ratones , Restricción Calórica , Leucemia Mieloide Aguda/patología , Histona Demetilasas/genética , Células Madre Neoplásicas/patología , Línea Celular Tumoral
2.
Cancer Rep (Hoboken) ; 6(1): e1625, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35546267

RESUMEN

BACKGROUND: Early-life stress due to poor parental care has been suggested to increase cancer risk, though, so far, no experimental evidence established a link between defective parental behavior and spontaneous tumorigenesis in progeny. Essential maternal behavior is regulated, in particular, by the oxytocin (OT) hormonal circuit, which in turn responds to stimuli from the offspring and impinges on the central nervous systems. METHODS: By providing L-368,899 OT receptor (OTR) inhibitor to lactating mothers, we set up a model of defective maternal care in p53 knockout mice. RESULTS: The progeny of these dams showed, later in life, higher cortisol levels, shortened life span and increased tumorigenic potential of bone marrow cells (BMC). Notably, these phenotypes were transmitted to the following generation. CONCLUSIONS: Therefore, the inhibition of OT function in mothers is a novel paradigm of early-life stress that is inherited across generations and increases cancer risk in tumor-prone mice.


Asunto(s)
Oxitocina , Estrés Psicológico , Animales , Femenino , Ratones , Carcinogénesis , Lactancia , Ratones Noqueados , Oxitocina/metabolismo , Oxitocina/farmacología , Proteína p53 Supresora de Tumor/genética
3.
Nutrients ; 14(13)2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35807850

RESUMEN

Metabolic regulation of cancer cell growth via AMP-activated protein kinase (AMPK) activation is a widely studied strategy for cancer treatment, including leukemias. Recent notions that naturally occurring compounds might have AMPK activity led to the search for nutraceuticals with potential AMPK-stimulating activity. We found that hydroxycitric acid (HCA), a natural, safe bioactive from the plant Garcinia gummi-gutta (cambogia), has potent AMPK activity in chronic myelogenous leukemia (CML) cell line K562. HCA is a known competitive inhibitor of ATP citrate lyase (ACLY) and is widely used as a weight loss inducer. We found that HCA was able to inhibit the growth of K562 cells in in vitro and in vivo xenograft models. At the mechanistic level, we identified a direct interaction between AMPK and ACLY that seems to be sensitive to HCA treatment. Additionally, HCA treatment resulted in the co-activation of AMPK and the mammalian target of rapamycin (mTOR) pathways. Moreover, we found an enhanced unfolded protein response as observed by activation of the eIF2α/ATF4 pathway that could explain the induction of cell cycle arrest at the G2/M phase and DNA fragmentation upon HCA treatment in K562 cells. Overall, these findings suggest HCA as a nutraceutical approach for the treatment of CMLs.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Leucemia Mielógena Crónica BCR-ABL Positiva , Proteínas Quinasas Activadas por AMP/metabolismo , Citratos/farmacología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Serina-Treonina Quinasas TOR
4.
Aging Cell ; 15(3): 446-54, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26968134

RESUMEN

Oxidative stress and telomere attrition are considered the driving factors of aging. As oxidative damage to telomeric DNA favors the erosion of chromosome ends and, in turn, telomere shortening increases the sensitivity to pro-oxidants, these two factors may trigger a detrimental vicious cycle. To check whether limiting oxidative stress slows down telomere shortening and related progeria, we have investigated the effect of p66SHC deletion, which has been shown to reduce oxidative stress and mitochondrial apoptosis, on late-generation TERC (telomerase RNA component)-deficient mice having short telomeres and reduced lifespan. Double mutant (TERC(-/-) p66SHC(-/-) ) mice were generated, and their telomere length, fertility, and lifespan investigated in different generations. Results revealed that p66SHC deletion partially rescues sterility and weight loss, as well as organ atrophy, of TERC-deficient mice, but not their short lifespan and telomere erosion. Therefore, our data suggest that p66SHC-mediated oxidative stress and telomere shortening synergize in some tissues (including testes) to accelerate aging; however, early mortality of late-generation mice seems to be independent of any link between p66SHC-mediated oxidative stress and telomere attrition.


Asunto(s)
Fertilidad/genética , Eliminación de Gen , Longevidad , Progeria/genética , Progeria/patología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Telomerasa/deficiencia , Animales , Daño del ADN , Genotipo , Ratones Endogámicos C57BL , Estrés Oxidativo , Fenotipo , ARN/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Análisis de Supervivencia , Telomerasa/metabolismo , Acortamiento del Telómero , Pérdida de Peso
5.
Aging Cell ; 11(1): 162-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22081964

RESUMEN

Deletion of the p66(Shc) gene results in lean and healthy mice, retards aging, and protects from aging-associated diseases, raising the question of why p66(Shc) has been selected, and what is its physiological role. We have investigated survival and reproduction of p66(Shc)-/- mice in a population living in a large outdoor enclosure for a year, subjected to food competition and exposed to winter temperatures. Under these conditions, deletion of p66(Shc) was strongly counterselected. Laboratory studies revealed that p66(Shc)-/- mice have defects in fat accumulation, thermoregulation, and reproduction, suggesting that p66(Shc) has been evolutionarily selected because of its role in energy metabolism. These findings imply that the health impact of targeting aging genes might depend on the specific energetic niche and caution should be exercised against premature conclusions regarding gene functions that have only been observed in protected laboratory conditions.


Asunto(s)
Envejecimiento/genética , Longevidad/genética , Proteínas Adaptadoras de la Señalización Shc/genética , Envejecimiento/metabolismo , Animales , Evolución Biológica , Regulación de la Temperatura Corporal/genética , Metabolismo Energético/genética , Femenino , Aptitud Genética/genética , Heterocigoto , Homocigoto , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Noqueados , Estaciones del Año , Proteínas Adaptadoras de la Señalización Shc/deficiencia , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
6.
Diabetes ; 59(9): 2306-14, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20566667

RESUMEN

OBJECTIVE: The redox enzyme p66Shc produces hydrogen peroxide and triggers proapoptotic signals. Genetic deletion of p66Shc prolongs life span and protects against oxidative stress. In the present study, we evaluated the role of p66Shc in an animal model of diabetic wound healing. RESEARCH DESIGN AND METHODS: Skin wounds were created in wild-type (WT) and p66Shc(-/-) control and streptozotocin-induced diabetic mice with or without hind limb ischemia. Wounds were assessed for collagen content, thickness and vascularity of granulation tissue, apoptosis, reepithelialization, and expression of c-myc and beta-catenin. Response to hind limb ischemia was also evaluated. RESULTS: Diabetes delayed wound healing in WT mice with reduced granulation tissue thickness and vascularity, increased apoptosis, epithelial expression of c-myc, and nuclear localization of beta-catenin. These nonhealing features were worsened by hind limb ischemia. Diabetes induced p66Shc expression and activation; wound healing was significantly faster in p66Shc(-/-) than in WT diabetic mice, with or without hind limb ischemia, at 1 and 3 months of diabetes duration and in both SV129 and C57BL/6 genetic backgrounds. Deletion of p66Shc reversed nonhealing features, with increased collagen content and granulation tissue thickness, and reduced apoptosis and expression of c-myc and beta-catenin. p66Shc deletion improved response to hind limb ischemia in diabetic mice in terms of tissue damage, capillary density, and perfusion. Migration of p66Shc(-/-) dermal fibroblasts in vitro was significantly faster than WT fibroblasts under both high glucose and hypoxia. CONCLUSIONS: p66Shc is involved in the delayed wound-healing process in the setting of diabetes and ischemia. Thus, p66Shc may represent a potential therapeutic target against this disabling diabetes complication.


Asunto(s)
Proteínas Adaptadoras de la Señalización Shc/metabolismo , Cicatrización de Heridas/fisiología , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Diabetes Mellitus/metabolismo , Diabetes Mellitus/fisiopatología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Lisina/análogos & derivados , Lisina/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Oxidación-Reducción , Proteínas Adaptadoras de la Señalización Shc/deficiencia , Proteínas Adaptadoras de la Señalización Shc/genética , Piel/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Tirosina/análogos & derivados , Tirosina/metabolismo , Cicatrización de Heridas/genética
7.
J Biol Chem ; 283(49): 34283-93, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-18838380

RESUMEN

Reactive oxygen species (ROS) and insulin signaling in the adipose tissue are critical determinants of aging and age-associated diseases. It is not clear, however, if they represent independent factors or they are mechanistically linked. We investigated the effects of ROS on insulin signaling using as model system the p66(Shc)-null mice. p66(Shc) is a redox enzyme that generates mitochondrial ROS and promotes aging in mammals. We report that insulin activates the redox enzyme activity of p66(Shc) specifically in adipocytes and that p66(Shc)-generated ROS regulate insulin signaling through multiple mechanisms, including AKT phosphorylation, Foxo localization, and regulation of selected insulin target genes. Deletion of p66(Shc) resulted in increased mitochondrial uncoupling and reduced triglyceride accumulation in adipocytes and in vivo increased metabolic rate and decreased fat mass and resistance to diet-induced obesity. In addition, p66(Shc-/-) mice showed impaired thermo-insulation. These findings demonstrate that p66(Shc)-generated ROS regulate the effect of insulin on the energetic metabolism in mice and suggest that intracellular oxidative stress might accelerate aging by favoring fat deposition and fat-related disorders.


Asunto(s)
Estrés Oxidativo , Proteínas Adaptadoras de la Señalización Shc/fisiología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Obesidad/genética , Oxidación-Reducción , Oxígeno/metabolismo , Fosforilación , Especies Reactivas de Oxígeno , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
8.
Endothelium ; 15(5-6): 276-87, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19065319

RESUMEN

Previous studies showed that p66(Shc-/-) mice on a very-high-fat diet (HFD) had reduced oxidative stress, foam cell, and early atherosclerotic lesion formation. Here, the authors have used hypercholesterolemic apolipoprotein E (ApoE(-/-)) mice to investigate the role of p66Shc deletion in advanced atheroma. The authors generated mice deficient of both ApoE and p66Shc genes (ApoE(-/-) /p66(Shc-/-)). They used microsatellite polymerase chain reaction (PCR) analysis to analyze the genetic background and considered only animals with a constant percentages of C57B6L and 129SV background strands (it was obtained the 50.3% +/- 6.4% of C57B6L background). Computer-assisted analysis revealed that advanced atherosclerotic lesions in ApoE(-/-)/p66(Shc+/+) were significantly larger than those observed in ApoE(-/-)/p66(Shc-/-). Accordingly, the lipid-laden macrophage foam cells and oxidation-specific epitopes in ApoE(-/-)/p66(shc+/+) HFD-treated groups were higher than those observed in normal diet (ND)-treated groups. Thus, p66(Shc-/-) plays an important protective role also against advanced atherosclerotic lesion formation. Finally, the authors have used microarray to investigate major changes in gene expression in aortas of mice with ApoE(-/-)/p66(Shc-/-) background treated with a very HFD in comparison to ApoE(-/-)/p66(Shc+/+) (these data have been confirmed by by real-time PCR and immunohistochemistry). DAVID (Database for Annotation, Visualization and Integrated Discovery) analysis revealed that CD36 antigen (CD36), tissue inhibitor of metalloproteinase 2 (TIMP2), apolipoprotein E (ApoE), acetyl-coenzyme A acetyltransferase 1 (ACAT1), and thrombospondin 1 (THBS1) can be involved in p66 deletion-dependent vascular protection through the adipocytokine/lipid signaling pathway.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/metabolismo , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Proteínas Adaptadoras de la Señalización Shc/genética , Adipoquinas/genética , Adipoquinas/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , Aorta/fisiopatología , Apolipoproteínas E/metabolismo , Aterosclerosis/fisiopatología , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Células Espumosas/metabolismo , Células Espumosas/patología , Alimentos Formulados/efectos adversos , Eliminación de Gen , Regulación de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Genotipo , Hipercolesterolemia/fisiopatología , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA