Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Genes Dev ; 24(13): 1403-17, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20595232

RESUMEN

The sterol regulatory element-binding protein (SREBP) transcription factor family is a critical regulator of lipid and sterol homeostasis in eukaryotes. In mammals, SREBPs are highly active in the fed state to promote the expression of lipogenic and cholesterogenic genes and facilitate fat storage. During fasting, SREBP-dependent lipid/cholesterol synthesis is rapidly diminished in the mouse liver; however, the mechanism has remained incompletely understood. Moreover, the evolutionary conservation of fasting regulation of SREBP-dependent programs of gene expression and control of lipid homeostasis has been unclear. We demonstrate here a conserved role for orthologs of the NAD(+)-dependent deacetylase SIRT1 in metazoans in down-regulation of SREBP orthologs during fasting, resulting in inhibition of lipid synthesis and fat storage. Our data reveal that SIRT1 can directly deacetylate SREBP, and modulation of SIRT1 activity results in changes in SREBP ubiquitination, protein stability, and target gene expression. In addition, chemical activators of SIRT1 inhibit SREBP target gene expression in vitro and in vivo, correlating with decreased hepatic lipid and cholesterol levels and attenuated liver steatosis in diet-induced and genetically obese mice. We conclude that SIRT1 orthologs play a critical role in controlling SREBP-dependent gene regulation governing lipid/cholesterol homeostasis in metazoans in response to fasting cues. These findings may have important biomedical implications for the treatment of metabolic disorders associated with aberrant lipid/cholesterol homeostasis, including metabolic syndrome and atherosclerosis.


Asunto(s)
Regulación hacia Abajo , Ayuno/fisiología , Sirtuina 1/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo , Acetilación , Animales , Benzamidas/farmacología , Caenorhabditis elegans , Línea Celular , Colesterol/biosíntesis , Regulación hacia Abajo/efectos de los fármacos , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Lípidos/biosíntesis , Ratones , Naftoles/farmacología , Niacinamida/farmacología , Estabilidad Proteica/efectos de los fármacos , Sirtuinas/antagonistas & inhibidores
2.
Nature ; 473(7346): 234-8, 2011 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-21499261

RESUMEN

Notch signalling is a key intercellular communication mechanism that is essential for cell specification and tissue patterning, and which coordinates critical steps of blood vessel growth. Although subtle alterations in Notch activity suffice to elicit profound differences in endothelial behaviour and blood vessel formation, little is known about the regulation and adaptation of endothelial Notch responses. Here we report that the NAD(+)-dependent deacetylase SIRT1 acts as an intrinsic negative modulator of Notch signalling in endothelial cells. We show that acetylation of the Notch1 intracellular domain (NICD) on conserved lysines controls the amplitude and duration of Notch responses by altering NICD protein turnover. SIRT1 associates with NICD and functions as a NICD deacetylase, which opposes the acetylation-induced NICD stabilization. Consequently, endothelial cells lacking SIRT1 activity are sensitized to Notch signalling, resulting in impaired growth, sprout elongation and enhanced Notch target gene expression in response to DLL4 stimulation, thereby promoting a non-sprouting, stalk-cell-like phenotype. In vivo, inactivation of Sirt1 in zebrafish and mice causes reduced vascular branching and density as a consequence of enhanced Notch signalling. Our findings identify reversible acetylation of the NICD as a molecular mechanism to adapt the dynamics of Notch signalling, and indicate that SIRT1 acts as rheostat to fine-tune endothelial Notch responses.


Asunto(s)
Células Endoteliales/enzimología , Regulación de la Expresión Génica , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Sirtuina 1/genética , Sirtuina 1/metabolismo , Acetilación , Animales , Células Endoteliales/citología , Técnicas de Inactivación de Genes , Silenciador del Gen , Células HEK293 , Humanos , Ratones , Mutación , Receptor Notch1/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
3.
Nature ; 450(7170): 712-6, 2007 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-18046409

RESUMEN

Calorie restriction extends lifespan and produces a metabolic profile desirable for treating diseases of ageing such as type 2 diabetes. SIRT1, an NAD+-dependent deacetylase, is a principal modulator of pathways downstream of calorie restriction that produce beneficial effects on glucose homeostasis and insulin sensitivity. Resveratrol, a polyphenolic SIRT1 activator, mimics the anti-ageing effects of calorie restriction in lower organisms and in mice fed a high-fat diet ameliorates insulin resistance, increases mitochondrial content, and prolongs survival. Here we describe the identification and characterization of small molecule activators of SIRT1 that are structurally unrelated to, and 1,000-fold more potent than, resveratrol. These compounds bind to the SIRT1 enzyme-peptide substrate complex at an allosteric site amino-terminal to the catalytic domain and lower the Michaelis constant for acetylated substrates. In diet-induced obese and genetically obese mice, these compounds improve insulin sensitivity, lower plasma glucose, and increase mitochondrial capacity. In Zucker fa/fa rats, hyperinsulinaemic-euglycaemic clamp studies demonstrate that SIRT1 activators improve whole-body glucose homeostasis and insulin sensitivity in adipose tissue, skeletal muscle and liver. Thus, SIRT1 activation is a promising new therapeutic approach for treating diseases of ageing such as type 2 diabetes.


Asunto(s)
Restricción Calórica , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Sirtuinas/agonistas , Acetilación , Sitio Alostérico , Animales , Glucemia/metabolismo , Dominio Catalítico , Línea Celular , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Drosophila melanogaster , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Insulina/metabolismo , Insulina/farmacología , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Resveratrol , Sirtuina 1 , Sirtuinas/metabolismo , Estilbenos/química , Estilbenos/farmacología
4.
Cardiovasc Res ; 89(2): 464-72, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20978007

RESUMEN

AIMS: The mammalian silent information regulator-two 1 (Sirt1) blunts the noxious effects of cardiovascular risk factors such as type 2 diabetes mellitus and obesity. Nevertheless, the role of Sirt1 in regulating the expression of tissue factor (TF), the key trigger of coagulation, and arterial thrombus formation remains unknown. METHODS AND RESULTS: Human as well as mouse cell lines were used for in vitro experiments, and C57Bl/6 mice for in vivo procedures. Sirt1 inhibition by splitomicin or sirtinol enhanced cytokine-induced endothelial TF protein expression as well as surface activity, while TF pathway inhibitor protein expression did not change. Sirt1 inhibition further enhanced TF mRNA expression, TF promoter activity, and nuclear translocation as well as DNA binding of the p65 subunit of nuclear factor-kappa B (NFκB/p65). Sirt1 siRNA enhanced TF protein and mRNA expression, and this effect was reduced in NFκB/p65(-/-) mouse embryonic fibroblasts reconstituted with non-acetylatable Lys(310)-mutant NFκB/p65. Activation of the mitogen-activated protein kinases p38, c-Jun NH(2)-terminal kinase, and p44/42 (ERK) remained unaffected. In vivo, mice treated with the Sirt1 inhibitor splitomicin exhibited enhanced TF activity in the arterial vessel wall and accelerated carotid artery thrombus formation in a photochemical injury model. CONCLUSION: We provide pharmacological and genetic evidence that Sirt1 inhibition enhances TF expression and activity by increasing NFκB/p65 activation in human endothelial cells. Furthermore, Sirt1 inhibition induces arterial thrombus formation in vivo. Hence, modulation of Sirt1 may offer novel therapeutic options for targeting thrombosis.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/metabolismo , Tromboplastina/metabolismo , Trombosis/etiología , Animales , Benzamidas/farmacología , Sitios de Unión , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células Endoteliales/enzimología , Activadores de Enzimas , Genes Reporteros , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Naftalenos/farmacología , Naftoles/farmacología , Regiones Promotoras Genéticas , Pironas/farmacología , Interferencia de ARN , ARN Mensajero/metabolismo , Resveratrol , Sirtuina 1/deficiencia , Sirtuina 1/genética , Estilbenos/farmacología , Tromboplastina/genética , Trombosis/sangre , Trombosis/enzimología , Trombosis/genética , Factor de Transcripción ReIA/deficiencia , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Transfección
5.
Sci Rep ; 1: 70, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22355589

RESUMEN

Sirt1 is an NAD(+)-dependent deacetylase that extends lifespan in lower organisms and improves metabolism and delays the onset of age-related diseases in mammals. Here we show that SRT1720, a synthetic compound that was identified for its ability to activate Sirt1 in vitro, extends both mean and maximum lifespan of adult mice fed a high-fat diet. This lifespan extension is accompanied by health benefits including reduced liver steatosis, increased insulin sensitivity, enhanced locomotor activity and normalization of gene expression profiles and markers of inflammation and apoptosis, all in the absence of any observable toxicity. Using a conditional SIRT1 knockout mouse and specific gene knockdowns we show SRT1720 affects mitochondrial respiration in a Sirt1- and PGC-1α-dependent manner. These findings indicate that SRT1720 has long-term benefits and demonstrate for the first time the feasibility of designing novel molecules that are safe and effective in promoting longevity and preventing multiple age-related diseases in mammals.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Longevidad/efectos de los fármacos , Obesidad/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Composición Corporal/efectos de los fármacos , Grasas de la Dieta/administración & dosificación , Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos
6.
J Biol Chem ; 284(36): 24394-405, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19535340

RESUMEN

SIRT3 is a major mitochondrial NAD(+)-dependent protein deacetylase playing important roles in regulating mitochondrial metabolism and energy production and has been linked to the beneficial effects of exercise and caloric restriction. SIRT3 is emerging as a potential therapeutic target to treat metabolic and neurological diseases. We report the first sets of crystal structures of human SIRT3, an apo-structure with no substrate, a structure with a peptide containing acetyl lysine of its natural substrate acetyl-CoA synthetase 2, a reaction intermediate structure trapped by a thioacetyl peptide, and a structure with the dethioacetylated peptide bound. These structures provide insights into the conformational changes induced by the two substrates required for the reaction, the acetylated substrate peptide and NAD(+). In addition, the binding study by isothermal titration calorimetry suggests that the acetylated peptide is the first substrate to bind to SIRT3, before NAD(+). These structures and biophysical studies provide key insight into the structural and functional relationship of the SIRT3 deacetylation activity.


Asunto(s)
Acetato CoA Ligasa/química , Proteínas Mitocondriales/química , NAD/química , Péptidos/química , Sirtuinas/química , Acetato CoA Ligasa/metabolismo , Acetilación , Humanos , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Péptidos/metabolismo , Unión Proteica/fisiología , Estructura Cuaternaria de Proteína , Sirtuina 3 , Sirtuinas/metabolismo , Relación Estructura-Actividad
7.
Protein Sci ; 18(3): 514-25, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19241369

RESUMEN

SIRT3 is a key mitochondrial protein deacetylase proposed to play key roles in regulating mitochondrial metabolism but there has been considerable debate about its actual size, the sequences required for activity, and its subcellular localization. A previously cloned mouse SIRT3 has high sequence similarity with the C-terminus of human SIRT3 but lacks an N-terminal mitochondrial targeting sequence and has no detectable deacetylation activity in vitro. Using 5' rapid amplification of cDNA ends, we cloned the entire sequence of mouse SIRT3, as well as rat and rabbit SIRT3. Importantly, we find that full-length SIRT3 protein localizes exclusively to the mitochondria, in contrast to reports of SIRT3 localization to the nucleus. We demonstrate that SIRT3 has no deacetylation activity in vitro unless the protein is truncated, consistent with human SIRT3. In addition, we determined the inhibition constants and mechanism of action for nicotinamide and a small molecule SIRT3 inhibitor against active mouse SIRT3 and show that the mechanisms are different for the two compounds with respect to peptide substrate and NAD(+). Thus, identification and characterization of the actual SIRT3 sequence should help resolve the debate about the nature of mouse SIRT3 and identify new mechanisms to modulate enzymatic activity.


Asunto(s)
Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Señales de Clasificación de Proteína , Sirtuinas/genética , Sirtuinas/metabolismo , Distribución Tisular/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Cultivadas , Clonación Molecular , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/química , Datos de Secuencia Molecular , Niacinamida/metabolismo , Conejos , Ratas , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Sirtuina 3 , Sirtuinas/antagonistas & inhibidores , Sirtuinas/química
8.
Biol Reprod ; 70(4): 1206-12, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14681204

RESUMEN

Ionizing radiation (IR) and consequent induction of DNA double-strand breaks (DSBs) causes activation of the protein ataxia telangiectasia mutated (ATM). Normally, ATM is present as inactive dimers; however, in response to DSBs, the ATM dimer partners cross-phosphorylate each other on serine 1981, and kinase active ATM monomers are subsequently released. We have studied the presence of both nonphosphorylated as well as active serine 1981 phosphorylated ATM (pS1981-ATM) in the mouse testis. In the nonirradiated testis, ATM was present in spermatogonia and spermatocytes until stage VII of the cycle of the seminiferous epithelium, whereas pS1981-ATM was found only to be present in the sex body of pachytene spermatocytes. In response to IR, ATM became activated by pS1981 cross-phosphorylation in spermatogonia and Sertoli cells. Despite the occurrence of endogenous programmed DSBs during the first meiotic prophase and the presence of ATM in both spermatogonia and spermatocytes, pS1981 phosphorylated ATM did not appear in spermatocytes after treatment with IR. These results show that spermatogonial ATM and ATM in the spermatocytes are differentially regulated. In the mitotically dividing spermatogonia, ATM is activated by cross-phosphorylation, whereas during meiosis nonphosphorylated ATM or differently phosphorylated ATM is already active. ATM has been shown to be present at the synapsed axes of the meiotic chromosomes, and in the ATM knock-out mice spermatogenesis stops at pachytene stage IV of the seminiferous epithelium, indicating that indeed nonphosphorylated ATM is functional during meiosis. Additionally, ATM is constitutively phosphorylated in the sex body where its continued presence remains an enigma.


Asunto(s)
Proteínas Serina-Treonina Quinasas/fisiología , Testículo/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Masculino , Meiosis , Ratones , Ratones Endogámicos , Ratones Noqueados , Fosforilación , Profase , Proteínas Serina-Treonina Quinasas/metabolismo , Espermatocitos/fisiología , Espermatogonias/fisiología , Testículo/efectos de la radiación , Distribución Tisular , Proteínas Supresoras de Tumor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA